Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Cancer Chemother Pharmacol ; 82(3): 407-418, 2018 09.
Article in English | MEDLINE | ID: mdl-29926131

ABSTRACT

PURPOSE: The phase I study characterized the safety, pharmacokinetics, anti-tumor activity, and recommended phase II dose/schedule of LY3164530 in patients with advanced or metastatic cancer. METHODS: Patients received LY3164530 on days 1 and 15 (Schedule 1: 300, 600, 1000, and 1250 mg) or Days 1, 8, 15, and 22 (Schedule 2: 500 and 600 mg) of each 28 days cycle. Dose escalation used a modified toxicity probability interval model. RESULTS: Dose escalation defined a maximum tolerated dose (MTD) of 1000 mg on Schedule 1 and 500 mg on Schedule 2. Treatment-emergent adverse events related to study treatment were consistent with epidermal growth factor receptor (EGFR) inhibition and included maculopapular rash/dermatitis acneiform (83%, Grade 3/4 17%), hypomagnesemia (55%, Grade 3/4 7%), paronychia (35%), fatigue (28%, Grade 3/4 3%), skin fissures (24%), and hypokalemia (21%, Grade 3/4 7%). Partial response was achieved in three patients on Schedule 2 with colorectal cancer (n = 2) or squamous cell cancer. Overall response rate (ORR) was 10.3%, disease control rate (ORR + stable disease [SD]) was 51.7 and 17.2% of patients had SD ≥ 4 months. The in vivo stability of the bispecific antibody was confirmed. Schedule 2 provided greater and more consistent inhibition of mesenchymal-epithelial transition (MET)/EGFR throughout the dosing interval than Schedule 1. CONCLUSIONS: Although this study defined the LY3164530 MTD and pharmacokinetics on both schedules, significant toxicities associated with EGFR inhibition and lack of a potential predictive biomarker limit future development. Nonetheless, the results provide insight into the development of bispecific antibody therapy.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/adverse effects , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Adult , Aged , Antibodies, Bispecific/blood , Biomarkers, Tumor/analysis , Dose-Response Relationship, Drug , Drug Administration Schedule , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/blood , ErbB Receptors/immunology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/blood , Neoplasms/enzymology , Neoplasms/pathology , Proto-Oncogene Proteins c-met/blood , Proto-Oncogene Proteins c-met/immunology
3.
Sci Rep ; 8(1): 3056, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445099

ABSTRACT

Anticancer efficacy of ginger phenolics (GPs) has been demonstrated in various in vitro assays and xenograft mouse models. However, only sub-therapeutic plasma concentrations of GPs were detected in human and mouse pharmacokinetic (PK) studies. Intriguingly, a significant portion of GPs occurred as phase II metabolites (mainly glucuronide conjugates) in plasma. To evaluate the disposition of GPs and understand the real players responsible for efficacy, we performed a PK and tissue distribution study in mice. Plasma exposure of GPs was similar on day 1 and 7, suggesting no induction or inhibition of clearance pathways. Both free and conjugated GPs accumulated in all tissues including tumors. While non-cytotoxicity of 6-ginerol glucuronide precluded the role of conjugated GPs in cell death, the free forms were cytotoxic against prostate cancer cells. The efficacy of ginger was best explained by the reconversion of conjugated GPs to free forms by ß-glucuronidase, which is over-expressed in the tumor tissue. This previously unrecognized two-step process suggests an instantaneous conversion of ingested free GPs into conjugated forms, followed by their subsequent absorption into systemic circulation and reconversion into free forms. This proposed model uncovers the mechanistic underpinnings of ginger's anticancer activity despite sub-therapeutic levels of free GPs in the plasma.


Subject(s)
Cell Line, Tumor/drug effects , Plant Extracts/pharmacology , Plant Extracts/pharmacokinetics , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Catechols/pharmacokinetics , Catechols/pharmacology , Cell Proliferation/drug effects , Zingiber officinale/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , PC-3 Cells , Phenols/pharmacokinetics , Phenols/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
4.
Mol Pharmacol ; 88(4): 768-78, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26240287

ABSTRACT

Radiotherapy, a therapeutic modality of cancer treatment, nonselectively damages normal tissues as well as tumor tissues. The search is ongoing for therapeutic agents that selectively reduce radiation-induced normal tissue injury without reducing tumoricidal effect, thereby increasing the therapeutic ratio of radiation therapy. Our laboratory established 5-(4-methylpiperazin-1-yl)-2-[2'-(3,4-dimethoxyphenyl)-5'benzimidazolyl] benzimidazole (DMA) as noncytotoxic radioprotector in mammalian cells. DMA showed an excellent radioprotection in mice at single nontoxic oral dose by a dose-reduction factor of 1.28. An oxygen radical absorbing capacity assay confirmed its free-radical quenching ability. Single bolus dose and 28-days of repeated administration of DMA in mice for toxicity studies determined an LD50 of >2000 mg/kg body weight (bw) and 225 mg/kg bw, respectively, suggesting DMA is safe. Histopathology, biochemical parameters, and relative organ weight analysis revealed insignificant changes in the DMA-treated animals. The pharmacokinetic study of DMA at oral and intravenous doses showed its C(max) = 1 hour, bioavailability of 8.84%, elimination half-life of 4 hours, and an enterohepatic recirculation. Biodistribution study in mice with Ehrlich ascites tumors showed that (99m)Tc-DMA achieved its highest concentration in 1 hour and was retained up to 4 hours in the lungs, liver, kidneys, and spleen, and in a low concentration in the tumor, a solicited property of any radioprotector to protect normal cells over cancerous cells. We observed that the single-dose treatment of tumor-bearing mice with DMA 2 hours before 8 Gy total body irradiation showed an impressive rescue of radiation-induced morbidity in terms of weight loss and mortality without a change in tumor response.


Subject(s)
Benzimidazoles/pharmacokinetics , Benzimidazoles/toxicity , Piperazines/pharmacokinetics , Piperazines/toxicity , Radiation-Protective Agents/pharmacokinetics , Radiation-Protective Agents/toxicity , Animals , Benzimidazoles/metabolism , Bisbenzimidazole/metabolism , Bisbenzimidazole/pharmacokinetics , Bisbenzimidazole/toxicity , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/metabolism , Carcinoma, Ehrlich Tumor/radiotherapy , Dose-Response Relationship, Radiation , Drug Evaluation, Preclinical/methods , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Piperazines/metabolism , Radiation-Protective Agents/metabolism , Survival Rate/trends , Tissue Distribution/drug effects , Tissue Distribution/physiology
5.
Eur J Pharm Sci ; 77: 90-9, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26026989

ABSTRACT

Noscapine (Nos), an antitussive benzylisoquinoline opium alkaloid, is a non-toxic tubulin-binding agent currently in Phase II clinical trials for cancer chemotherapy. While preclinical studies have established its tumor-inhibitory properties in various cancers, poor absorptivity and rapid first-pass metabolism producing several uncharacterized metabolites for efficacy, present an impediment in translating its efficacy in humans. Here we report novel formulations of Nos in combination with dietary agents like capsaicin (Cap), piperine (Pip), eugenol (Eu) and curcumin (Cur) known for modulating Phase I and II drug metabolizing enzymes. In vivo pharmacokinetic (PK), organ toxicity evaluation of combinations, microsomal stability and in vitro cytochrome P450 (CYP) inhibition effects of Nos, Cap and Pip using human liver microsomes were performed. Single-dose PK screening of combinations revealed that the relative exposure of Nos (2 µg h/mL) was enhanced by 2-fold (4 µg h/mL) by Cap and Pip and their plasma concentration-time profiles showed multiple peaking phenomena for Nos indicating enterohepatic recirculation or differential absorption from intestine. CYP inhibition studies confirmed that Nos, Cap and Pip are not potent CYP inhibitors (IC50>1 µM). Repeated oral dosing of Nos, Nos+Cap and Nos+Pip showed lower exposure (Cmax and AUClast) of Nos on day 7 compared to day 1. Nos Cmax decreased from 3087 ng/mL to 684 ng/mL and AUClast from 1024 ng h/mL to 508 ng h/mL. In presence of Cap and Pip, the decrease in Cmax and AUClast of Nos was similar. This may be due to potential enzyme induction leading to rapid clearance of Nos as the trend was observed in Nos alone group also. The lack of effect on intrinsic clearance of Nos suggests that the potential drug biotransformation modulators employed in this study did not contribute toward increased exposure of Nos on repeated dosing. We envision that Nos-induced enzyme induction could alter the therapeutic efficacy of co-administered drugs, hence emphasizing the need for strategic evaluation of the metabolism of Nos to reap its maximum efficacy.


Subject(s)
Antitussive Agents/pharmacokinetics , Intestinal Mucosa/metabolism , Liver/metabolism , Noscapine/pharmacokinetics , Animals , Food-Drug Interactions , Humans , Male , Mice
6.
Bioorg Med Chem Lett ; 25(10): 2133-40, 2015.
Article in English | MEDLINE | ID: mdl-25891106

ABSTRACT

Noscapine is an opium-derived kinder-gentler microtubule-modulating drug, currently in Phase I/II clinical trials for cancer chemotherapy. Here, we report the synthesis of four more potent di-substituted brominated derivatives of noscapine, 9-Br-7-OH-NOS (2), 9-Br-7-OCONHEt-NOS (3), 9-Br-7-OCONHBn-NOS (4), and 9-Br-7-OAc-NOS (5) and their chemotherapeutic efficacy on PC-3 and MDA-MB-231 cells. The four derivatives were observed to have higher tubulin binding activity than noscapine and significantly affect tubulin polymerization. The equilibrium dissociation constant (KD) for the interaction between tubulin and 2, 3, 4, 5 was found to be, 55±6µM, 44±6µM, 26±3µM, and 21±1µM respectively, which is comparable to parent analog. The effects of these di-substituted noscapine analogs on cell cycle parameters indicate that the cells enter a quiescent phase without undergoing further cell division. The varying biological activity of these analogs and bulk of substituent at position-7 of the benzofuranone ring system of the parent molecule was rationalized utilizing predictive in silico molecular modeling. Furthermore, the immunoblot analysis of protein lysates from cells treated with 4 and 5, revealed the induction of apoptosis and down-regulation of survivin levels. This result was further supported by the enhanced activity of caspase-3/7 enzymes in treated samples compared to the controls. Hence, these compounds showed a great potential for studying microtubule-mediated processes and as chemotherapeutic agents for the management of human cancers.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Design , Microtubules/drug effects , Noscapine/chemical synthesis , Noscapine/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Models, Molecular , Noscapine/chemistry
7.
Mol Pharm ; 11(12): 4339-49, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25350222

ABSTRACT

Here, we report improved solubility and enhanced colonic delivery of reduced bromonoscapine (Red-Br-Nos), a cyclic ether brominated analogue of noscapine, upon encapsulation of its cyclodextrin (CD) complexes in bioresponsive guar gum microspheres (GGM). Phase-solubility analysis suggested that Red-Br-Nos complexed with ß-CD and methyl-ß-CD in a 1:1 stoichiometry, with a stability constant (Kc) of 2.29 × 10(3) M(-1) and 4.27 × 10(3) M(-1). Fourier transforms infrared spectroscopy indicated entrance of an O-CH2 or OCH3-C6H4-OCH3 moiety of Red-Br-Nos in the ß-CD or methyl-ß-CD cavity. Furthermore, the cage complex of Red-Br-Nos with ß-CD and methyl-ß-CD was validated by several spectral techniques. Rotating frame Overhauser enhancement spectroscopy revealed that the Ha proton of the OCH3-C6H4-OCH3 moiety was closer to the H5 proton of ß-CD and the H3 proton of the methyl-ß-CD cavity. The solubility of Red-Br-Nos in phosphate buffer saline (PBS, pH ∼ 7.4) was improved by ∼10.7-fold and ∼21.2-fold when mixed with ß-CD and methyl-ß-CD, respectively. This increase in solubility led to a favorable decline in the IC50 by ∼2-fold and ∼3-fold for Red-Br-Nos-ß-CD-GGM and Red-Br-Nos-methyl-ß-CD-GGM formulations respectively, compared to free Red-Br-Nos-ß-CD and Red-Br-Nos-methyl-ß-CD in human colon HT-29 cells. GGM-bearing drug complex formulations were found to be highly cytotoxic to the HT-29 cell line and further effective with simultaneous continuous release of Red-Br-Nos from microspheres. This is the first study to showing the preparation of drug-complex loaded GGMS for colon delivery of Red-Br-Nos that warrants preclinical assessment for the effective management of colon cancer.


Subject(s)
Cyclodextrins/chemistry , Galactans/chemistry , Mannans/chemistry , Microspheres , Noscapine/chemistry , Plant Gums/chemistry , Calorimetry, Differential Scanning , HT29 Cells , Humans , Spectroscopy, Fourier Transform Infrared , beta-Cyclodextrins/chemistry
8.
PLoS One ; 9(9): e108386, 2014.
Article in English | MEDLINE | ID: mdl-25251219

ABSTRACT

Natural and complementary therapies in conjunction with mainstream cancer care are steadily gaining popularity. Ginger extract (GE) confers significant health-promoting benefits owing to complex additive and/or synergistic interactions between its bioactive constituents. Recently, we showed that preservation of natural "milieu" confers superior anticancer activity on GE over its constituent phytochemicals, 6-gingerol (6G), 8-gingerol (8 G), 10-gingerol (10 G) and 6-shogaol (6S), through enterohepatic recirculation. Here we further evaluate and compare the effects of GE and its major bioactive constituents on cytochrome P450 (CYP) enzyme activity in human liver microsomes by monitoring metabolites of CYP-specific substrates using LC/MS/MS detection methods. Our data demonstrate that individual gingerols are potent inhibitors of CYP isozymes, whereas GE exhibits a much higher half-maximal inhibition value, indicating no possible herb-drug interactions. However, GE's inhibition of CYP1A2 and CYP2C8 reflects additive interactions among the constituents. In addition, studies performed to evaluate transporter-mediated intestinal efflux using Caco-2 cells revealed that GE and its phenolics are not substrates of P-glycoprotein (Pgp). Intriguingly, however, 10 G and 6S were not detected in the receiver compartment, indicating possible biotransformation across the Caco-2 monolayer. These data strengthen the notion that an interplay of complex interactions among ginger phytochemicals when fed as whole extract dictates its bioactivity highlighting the importance of consuming whole foods over single agents. Our study substantiates the need for an in-depth analysis of hepatic biotransformation events and distribution profiles of GE and its active phenolics for the design of safe regimens.


Subject(s)
Catechols/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Microsomes, Liver/enzymology , Plant Extracts/pharmacology , Zingiber officinale/chemistry , Caco-2 Cells , Catechols/chemistry , Cytochrome P-450 Enzyme Inhibitors/chemistry , Cytochrome P-450 Enzyme System/metabolism , Fatty Alcohols/pharmacology , Gene Expression Regulation/drug effects , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Microsomes, Liver/drug effects , Plant Extracts/chemistry
9.
Biochem Pharmacol ; 92(2): 192-205, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25124704

ABSTRACT

Noscapine, an opium-derived 'kinder-gentler' microtubule-modulating drug is in Phase I/II clinical trials for cancer chemotherapy. However, its limited water solubility encumbers its development into an oral anticancer drug with clinical promise. Here we report the synthesis of 9 third-generation, water-soluble noscapine analogs with negatively charged sulfonato and positively charged quaternary ammonium groups using noscapine, 9-bromonoscapine and 9-aminonoscapine as scaffolds. The predictive free energy of solvation was found to be lower for sulfonates (6a-c; 8a-c) compared to the quaternary ammonium-substituted counterparts, explaining their higher water solubility. In addition, sulfonates showed higher charge dispersability, which may effectively shield the hydrophobicity of isoquinoline nucleus as indicated by hydrophobicity mapping methods. These in silico data underscore efficient net charge balancing, which may explain higher water solubility and thus enhanced antiproliferative efficacy and improved bioavailability. We observed that 6b, 8b and 8c strongly inhibited tubulin polymerization and demonstrated significant antiproliferative activity against four cancer cell lines compared to noscapine. Molecular simulation and docking studies of tubulin-drug complexes revealed that the brominated compound with a four-carbon chain (4b, 6b, and 8b) showed optimal binding with tubulin heterodimers. Interestingly, 6b, 8b and 8c treated PC-3 cells resulted in preponderance of mitotic cells with multipolar spindle morphology, suggesting that they stall the cell cycle. Furthermore, in vivo pharmacokinetic evaluation of 6b, 8b and 8c revealed at least 1-2-fold improvement in their bioavailability compared to noscapine. To our knowledge, this is the first report to demonstrate novel water-soluble noscapine analogs that may pave the way for future pre-clinical drug development.


Subject(s)
Cell Proliferation/drug effects , Growth Inhibitors/pharmacology , Microtubules/drug effects , Noscapine/analogs & derivatives , Tubulin Modulators/pharmacology , Water/chemistry , Animals , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Goats , Growth Inhibitors/chemistry , HeLa Cells , Humans , Male , Mice , Microtubules/physiology , Noscapine/pharmacology , Protein Structure, Secondary , Solubility , Tubulin Modulators/chemistry
10.
Anticancer Drugs ; 25(6): 704-16, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24642711

ABSTRACT

Noscapine (Nos), an orally available plant-derived antitussive alkaloid, is in phase II clinical trials for cancer chemotherapy. It has extensively been shown to inhibit tumor growth in nude mice bearing human xenografts of hematopoietic, breast, lung, ovarian, brain, and prostate origin. However, high tumor-suppressive Nos dosages encumber the development of oral controlled-release formulations because of a short biological half-life (<2 h), poor absorption, low aqueous solubility, and extensive first-pass metabolism. Here, we present the design, fabrication, optimization, characterization, and biological evaluation of estrone-conjugated noscapine-loaded gelatin nanoparticles (Nos-ES-GN) for targeting estrogen-receptor-positive breast cancer MCF-7 cells. Gelatin nanoparticles (GN) were a uniformly compact size, stable at physiological pH, and showed a drug entrapment efficiency of 66.1±5.9 and 65.2±5.6% for Nos-GN and Nos-ES-GN, respectively. The secondary structure of gelatin nanocoacervates was predicted using circular dichroism and in-silico molecular modeling. Our data suggest that ethanol-fabricated GN retained the α-helical content of gelatin, whereas acetone favored the formation of random coils. The conjugation of estrone to Nos-GN did not affect the release rate of the drug, and both formulations followed first-order release kinetics with an initial burst, followed by a slow release. The IC50 value of Nos-ES-GN was 21.2 µmol/l, which was ∼50% lower than the free drug (43.3 µmol/l), suggesting targeted drug delivery. Our cell uptake study carried out in an estrogen-receptor-positive (MCF-7) and negative (MDA-MB-231) cancer cell lines showed greater accumulation of Nos-ES-GN in MCF-7 cells instead of MDA-MB-231 cells. Our data indicated that estrone-conjugated nanoparticles may potentially be used for targeting breast cancer cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Antitussive Agents/administration & dosage , Noscapine/administration & dosage , Receptors, Estrogen/metabolism , Antineoplastic Agents/chemistry , Antitussive Agents/chemistry , Breast Neoplasms/drug therapy , Cell Line, Tumor , Drug Carriers , Drug Delivery Systems , Estrone/chemistry , Female , Gelatin , Humans , Inhibitory Concentration 50 , Molecular Targeted Therapy , Nanoparticles , Noscapine/chemistry , Protein Structure, Secondary
11.
Cancer Prev Res (Phila) ; 7(5): 477-86, 2014 May.
Article in English | MEDLINE | ID: mdl-24449055

ABSTRACT

Plants contain a much greater diversity of bioactive compounds than any man-made chemical library. Heart-shaped Piper betel leaves are magnificent reservoirs of phenolic compounds with antiproliferative, antimutagenic, antibacterial, and antioxidant properties. Widely consumed in South Asian countries, the glossy leaf contains a multitude of biophenolics such as hydroxychavicol, eugenol, chavibetol, and piperols. Convincing data underscore the remarkable chemotherapeutic and chemopreventive potential of betel leaves against a variety of cancer types. The leaf constituents modulate an extensive array of signaling molecules such as transcription factors as well as reactive oxygen species (ROS) to control multiple nodes of various cellular proliferation and death pathways. Herein, we provide an overall perspective on the cancer-fighting benefits of the phenolic phytochemicals in betel leaves and a comprehensive overview of the mechanisms responsive to dose-driven ROS-mediated signaling cascades conscripted by bioactive phenolics to confer chemotherapeutic and chemopreventive advantages. Intriguingly, these ROS-triggered responses are contextual and may either elicit a protective xenohormetic antioxidant response to premalignant cells to constitute a chemopreventive effect or generate a curative chemotherapeutic response by pro-oxidatively augmenting the constitutively elevated ROS levels in cancer cells to tip the balance in favor of selective apoptosis induction in cancer cells while sparing normal ones. In conclusion, this review provides an update on how distinct ROS levels exist in normal versus cancer cells and how these levels can be strategically modulated and exploited for therapeutic gains. We emphasize the yet untapped potential of the evergreen vine, betel leaf, for chemopreventive and chemotherapeutic management of cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Dietary Supplements , Neoplasms/drug therapy , Piper betle , Plant Extracts/therapeutic use , Plant Leaves , Animals , Antineoplastic Agents, Phytogenic/chemistry , Chemoprevention/methods , Hormesis , Humans , Neoplasms/prevention & control , Phytotherapy , Piper betle/chemistry , Plant Leaves/chemistry , Reactive Oxygen Species/metabolism
12.
Carcinogenesis ; 35(6): 1320-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24431413

ABSTRACT

Phytochemical complexity of plant foods confers health-promoting benefits including chemopreventive and anticancer effects. Isolating single constituents from complex foods may render them inactive, emphasizing the importance of preserving the natural composition of whole extracts. Recently, we demonstrated in vitro synergy among the most abundant bioactive constituents of ginger extract (GE), viz., 6-gingerol (6G), 8-gingerol (8G), 10-gingerol (10G) and 6-shogaol (6S). However, no study has yet examined the in vivo collaboration among ginger phytochemicals or evaluated the importance, if any, of the natural 'milieu' preserved in whole extract. Here, we comparatively evaluated in vivo efficacy of GE with an artificial quasi-mixture (Mix) formulated by combining four most active ginger constituents at concentrations equivalent to those present in whole extract. Orally fed GE showed 2.4-fold higher tumor growth-inhibitory efficacy than Mix in human prostate tumor xenografts. Pharmacokinetic evaluations and bioavailability measurements addressed the efficacy differences between GE and Mix. Plasma concentration-time profiles revealed multiple peaking phenomenon for ginger constituents when they were fed as GE as opposed to Mix, indicating enterohepatic recirculation. Bioavailability of 6G, 8G, 10G and 6S was 1.6-, 1.1-, 2.5- and 3.4-fold higher, respectively, when dosed with GE compared with Mix. In addition, gingerol glucuronides were detected in feces upon intravenous administration confirming hepatobiliary elimination. These data ascribe the superior in vivo efficacy of GE to higher area under the concentration time curves, greater residence time and enhanced bioavailability, of ginger phytochemicals, when fed as a natural extract compared with artificial Mix, emphasizing the usefulness of consuming whole foods over single agents.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Enterohepatic Circulation , Phytochemicals/pharmacology , Plant Extracts/pharmacology , Zingiber officinale/chemistry , Administration, Intravenous , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Catechols/chemistry , Catechols/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Fatty Alcohols/chemistry , Fatty Alcohols/metabolism , Humans , Male , Mice , Phenols/chemistry , Phytochemicals/administration & dosage , Phytochemicals/pharmacokinetics , Plant Extracts/administration & dosage , Plant Extracts/pharmacokinetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Carcinogenesis ; 34(9): 2039-49, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23629419

ABSTRACT

Polyphenolic phytochemicals present in fruits and vegetables indisputably confer anticancer benefits upon regular consumption. Recently, we demonstrated the growth-inhibitory and apoptosis-inducing properties of polyphenol-rich sweet potato greens extract (SPGE) in cell culture and in vivo prostate cancer xenograft models. However, the bioactive constituents remain elusive. Here, we report a bioactivity-guided fractionation of SPGE based upon differential solvent polarity using chromatographic techniques that led to the identification of a remarkably active polyphenol-enriched fraction, F5, which was ~100-fold more potent than the parent extract as shown by IC50 measurements in human prostate cancer cells. High-performance liquid chromatography-ultraviolet and mass spectrometric analyses of the seven SPGE fractions suggested varying abundance of the major phenols, quinic acid (QA), caffeic acid, its ester chlorogenic acid, and isochlorogenic acids, 4,5-di-CQA, 3,5-di-CQA and 3,4-di-CQA, with a distinct composition of the most active fraction, F5. Subfractionation of F5 resulted in loss of bioactivity, suggesting synergistic interactions among the constituent phytochemicals. Quantitative analyses revealed a ~2.6- and ~3.6-fold enrichment of QA and chlorogenic acid, respectively, in F5 and a definitive ratiometric relationship between the isochlorogenic acids. Daily oral administration of 400mg/kg body wt of F5 inhibited growth and progression of prostate tumor xenografts by ~75% in nude mice, as evidenced by tumor volume measurements and non-invasive real-time bioluminescence imaging. These data generate compelling grounds to further examine the chemopreventive efficacy of the most active fraction of SPGE and suggest its potential usefulness as a dietary supplement for prostate cancer management.


Subject(s)
Cell Proliferation/drug effects , Ipomoea batatas/chemistry , Plant Extracts/administration & dosage , Polyphenols/administration & dosage , Prostatic Neoplasms/diet therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Chromatography, High Pressure Liquid , Dietary Supplements , Drug Synergism , Humans , Male , Mice , Plant Extracts/chemistry , Polyphenols/chemistry , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
14.
Nutr Cancer ; 65(2): 263-72, 2013.
Article in English | MEDLINE | ID: mdl-23441614

ABSTRACT

Dietary phytochemicals offer nontoxic therapeutic management as well as chemopreventive intervention for slow-growing prostate cancers. However, the limited success of several single-agent clinical trials suggest a paradigm shift that the health benefits of fruits and vegetables are not ascribable to individual phytochemicals, rather may be ascribed to synergistic interactions among them. We recently reported growth-inhibiting and apoptosis-inducing properties of ginger extract (GE) in in vitro and in vivo prostate cancer models. Nevertheless, the nature of interactions among the constituent ginger biophenolics, viz. 6-gingerol, 8-gingerol, 10-gingerol, and 6-shogoal, remains elusive. Here we show antiproliferative efficacy of the most-active GE biophenolics as single-agents and in binary combinations, and investigate the nature of their interactions using the Chou-Talalay combination index (CI) method. Our data demonstrate that binary combinations of ginger phytochemicals synergistically inhibit proliferation of PC-3 cells with CI values ranging from 0.03 to 0.88. To appreciate synergy among phytochemicals present in GE, the natural abundance of ginger biophenolics was quantitated using LC-UV/MS. Interestingly, combining GE with its constituents (in particular, 6-gingerol) resulted in significant augmentation of GE's antiproliferative activity. These data generate compelling grounds for further preclinical evaluation of GE alone and in combination with individual ginger biophenols for prostate cancer management.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Catechols/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Zingiber officinale/chemistry , Catechols/chemistry , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid/methods , Drug Synergism , Fatty Alcohols/pharmacology , Humans , Male , Phytochemicals/analysis , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/analysis , Plant Extracts/chemistry , Plant Extracts/pharmacology
15.
Carcinogenesis ; 34(7): 1558-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23430955

ABSTRACT

Plant extracts, a concoction of bioactive non-nutrient phytochemicals, have long served as the most significant source of new leads for anticancer drug development. Explored for their unique medicinal properties, the leaves of Piper betel, an evergreen perennial vine, are a reservoir of phenolics with antimutagenic, antitumor and antioxidant activities. Here, we show that oral feeding of betel leaf extract (BLE) significantly inhibited the growth of human prostate xenografts implanted in nude mice compared with vehicle-fed controls. To gain insights into the 'active principles', we performed a bioactivity-guided fractionation of methanolic BLE employing solvents of different polarity strengths using classical column chromatography. This approach yielded 15 fractions, which were then pooled to 10 using similar retention factors on thin-layer chromatographs. Bioactivity assays demonstrated that one fraction in particular, F2, displayed a 3-fold better in vitro efficacy to inhibit proliferation of prostate cancer cells than the parent BLE. The presence of phenols, hydroxychavicol (HC) and chavibetol (CHV), was confirmed in F2 by nuclear magnetic resonance, high-performance liquid chromatography and mass spectroscopy. Further, the HC containing F2 subfraction was found to be ~8-fold more potent than the F2 subfraction that contained CHV, in human prostate cancer PC-3 cells as evaluated by the 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide assay. Removing CHV from F2 remarkably decreased the IC50 of this fraction, indicating that HC is perhaps the major bioactive constituent, which is present to an extent of 26.59% in BLE. These data provide evidence that HC is a potential candidate for prostate cancer management and warrants further preclinical evaluation.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Piper/chemistry , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Chemical Fractionation , Chromatography, High Pressure Liquid , Eugenol/analogs & derivatives , Eugenol/chemistry , Eugenol/isolation & purification , Eugenol/therapeutic use , Humans , Immunohistochemistry , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Male , Methanol/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Solvents/chemistry , Xenograft Model Antitumor Assays
16.
Br J Nutr ; 107(4): 473-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21849094

ABSTRACT

It is appreciated far and wide that increased and regular consumption of fruits and vegetables is linked with noteworthy anticancer benefits. Extensively consumed as a spice in foods and beverages worldwide, ginger (Zingiber officinale Roscoe) is an excellent source of several bioactive phenolics, including non-volatile pungent compounds such as gingerols, paradols, shogaols and gingerones. Ginger has been known to display anti-inflammatory, antioxidant and antiproliferative activities, indicating its promising role as a chemopreventive agent. Here, we show that whole ginger extract (GE) exerts significant growth-inhibitory and death-inductory effects in a spectrum of prostate cancer cells. Comprehensive studies have confirmed that GE perturbed cell-cycle progression, impaired reproductive capacity, modulated cell-cycle and apoptosis regulatory molecules and induced a caspase-driven, mitochondrially mediated apoptosis in human prostate cancer cells. Remarkably, daily oral feeding of 100 mg/kg body weight of GE inhibited growth and progression of PC-3 xenografts by approximately 56 % in nude mice, as shown by measurements of tumour volume. Tumour tissue from GE-treated mice showed reduced proliferation index and widespread apoptosis compared with controls, as determined by immunoblotting and immunohistochemical methods. Most importantly, GE did not exert any detectable toxicity in normal, rapidly dividing tissues such as gut and bone marrow. To the best of our knowledge, this is the first report to demonstrate the in vitro and in vivo anticancer activity of whole GE for the management of prostate cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Dietary Supplements , Plant Extracts/therapeutic use , Prostatic Neoplasms/diet therapy , Rhizome/chemistry , Zingiber officinale/chemistry , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/metabolism , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Biomarkers/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Dietary Supplements/adverse effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Plant Extracts/adverse effects , Plant Extracts/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/prevention & control , Tumor Burden , Xenograft Model Antitumor Assays
17.
Carcinogenesis ; 32(12): 1872-80, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21948980

ABSTRACT

Sweet potato (Ipomoea batatas) leaves or greens, extensively consumed as a vegetable in Africa and Asia, are an excellent source of dietary polyphenols such as anthocyanins and phenolic acids. Here, we show that sweet potato greens extract (SPGE) has the maximum polyphenol content compared with several commercial vegetables including spinach. The polyphenol-rich SPGE exerts significant antiproliferative activity in a panel of prostate cancer cell lines while sparing normal prostate epithelial cells. Mechanistically, SPGE perturbed cell cycle progression, reduced clonogenic survival, modulated cell cycle and apoptosis regulatory molecules and induced apoptosis in human prostate cancer PC-3 cells both in vitro and in vivo. SPGE-induced apoptosis has a mitochondrially mediated component, which was attenuated by pretreatment with cyclosporin A. We also observed alterations of apoptosis regulatory molecules such as inactivation of Bcl2, upregulation of BAX, cytochrome c release and activation of downstream apoptotic signaling. SPGE caused DNA degradation as evident by terminal deoxynucleotidyl transferase-mediated dUTP-nick-end labeling (TUNEL) staining of increased concentration of 3'-DNA ends. Furthermore, apoptotic induction was caspase dependent as shown by cleavage of caspase substrate, poly (adenosine diphosphate-ribose) polymerase. Oral administration of 400 mg/kg SPGE remarkably inhibited growth and progression of prostate tumor xenografts by ∼69% in nude mice, as shown by tumor volume measurements and non-invasive real-time bioluminescent imaging. Most importantly, SPGE did not cause any detectable toxicity to rapidly dividing normal tissues such as gut and bone marrow. This is the first report to demonstrate the in vitro and in vivo anticancer activity of sweet potato greens in prostate cancer.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Ipomoea batatas/chemistry , Plant Leaves/chemistry , Polyphenols/pharmacology , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Nude , Microscopy, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...