Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Biomed Pharmacother ; 130: 110539, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32768880

ABSTRACT

Side effects of chemotherapy are burning questions for physicians and patients involved in cancers. Ganoderma lucidum is a widely consumed traditional Chinese medicine and edible mushroom with multiple functional properties. The present study aims to investigate the potential of polysaccharides from spore of G. lucidum (SGP) on small intestinal barrier function recovery against paclitaxel (PTX) challenge in a breast cancer mice model and IEC-6 cell line. The 4T1 tumor-bearing mice were treated with PTX together with four-week daily oral administration of SGP. Results indicated that combination of PTX and SGP reversed body weight lost and remolded the histology of small intestine, accompanied with promoted proliferation but suppressed apoptosis in intestinal cells. Intestinal barrier function was enhanced by the combination as indicated by reduced endotoxemia and the up-regulation of tight junction proteins, including Zonula occludens-1 (ZO-1), E-cadherin, ß-catenin and Occludin. The protection of SGP was further confirmed in IEC-6 cells affected by PTX in vitro. The combination treatment prevented PTX-induced apoptosis in IEC-6 by inhibiting microtubule polymerization, and the aforementioned tight junction proteins were also upregulated. These findings suggest a promising protective effect of SGP against small intestinal barrier injury caused by PTX, highlighting its clinical implication against the chemotherapy side effects.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Apoptosis/drug effects , Intestinal Mucosa/drug effects , Microtubules/drug effects , Paclitaxel/toxicity , Polysaccharides/pharmacology , Reishi/chemistry , Spores, Fungal/chemistry , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Female , Humans , Intestinal Mucosa/cytology , Mice , Mice, Inbred BALB C , Paclitaxel/antagonists & inhibitors , Polysaccharides/chemistry , Tight Junction Proteins/biosynthesis , Weight Loss/drug effects
2.
Biomed Pharmacother ; 129: 110441, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32580047

ABSTRACT

Caffeine (1,3,7-trimethylxanthine) is a xanthine alkaloid found in a number of dietary products consumed worldwide, such as coffee, tea, and soft beverages, and is known to act as a modifying agent for cytotoxic chemotherapeutic drugs. Studies have shown that caffeine reduces the cytotoxic effects of paclitaxel and inhibits paclitaxel-induced apoptosis; however, the underlying mechanism remains unclear. Here, we investigated whether caffeine inhibits the antitumor activity of paclitaxel via down-regulation of α-tubulin acetylation. In vitro studies, involving MTT assay, wound-healing assay, cell apoptosis assay, and western blotting analysis of A549 and HeLa cells, were performed. A549 and HeLa cell-based xenografts were established, and western blotting and immunohistochemical staining were performed for in vivo studies. The results showed that caffeine promoted the growth of cancer cells treated with paclitaxel. Additionally, caffeine enhanced migration ability, inhibited apoptosis, and decreased the acetylation of α-tubulin in paclitaxel-treated cancer cells. Furthermore, caffeine decreased the inhibitory effect of paclitaxel on tumor growth through down-regulation of α-tubulin acetylation in vivo. Taken together, these findings demonstrate that caffeine inhibits the anticancer activity of paclitaxel via down-regulation of α-tubulin acetylation, suggesting that patients receiving treatment with taxanes, such as paclitaxel, should avoid consuming caffeinated beverages or foods.


Subject(s)
Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Caffeine/pharmacology , Lung Neoplasms/drug therapy , Paclitaxel/antagonists & inhibitors , Tubulin/metabolism , Uterine Cervical Neoplasms/drug therapy , A549 Cells , Acetylation , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Interactions , Female , HeLa Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Paclitaxel/pharmacology , Protein Processing, Post-Translational , Signal Transduction , Tumor Burden/drug effects , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
Bull Exp Biol Med ; 166(2): 217-221, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30488215
4.
Neuropharmacology ; 141: 201-213, 2018 10.
Article in English | MEDLINE | ID: mdl-30179591

ABSTRACT

Given our recent evidence for the role of high mobility group box 1 (HMGB1) in chemotherapy-induced peripheral neuropathy (CIPN) in rats, we examined the origin of HMGB1 and the upstream and downstream mechanisms of HMGB1 release involved in paclitaxel-induced neuropathy in mice. Paclitaxel treatment developed mechanical allodynia in mice, as assessed by von Frey test, which was prevented by an anti-HMGB1-neutralizing antibody or thrombomodulin alfa capable of inactivating HMGB1. RAGE or CXCR4 antagonists, ethyl pyruvate or minocycline, known to inhibit HMGB1 release from macrophages, and liposomal clodronate, a macrophage depletor, prevented the paclitaxel-induced allodynia. Paclitaxel caused upregulation of RAGE and CXCR4 in the dorsal root ganglia and macrophage accumulation in the sciatic nerve. In macrophage-like RAW264.7 cells, paclitaxel evoked cytoplasmic translocation of nuclear HMGB1 followed by its extracellular release, and overexpression of CBP and PCAF, histone acetyltransferases (HATs), known to cause acetylation and cytoplasmic translocation of HMGB1, which were suppressed by ethyl pyruvate, N-acetyl-l-cysteine, an anti-oxidant, and SB203580 and PDTC, inhibitors of p38 MAP kinase (p38MAPK) and NF-κB, respectively. Paclitaxel increased accumulation of reactive oxygen species (ROS) and phosphorylation of p38MAPK, NF-κB p65 and I-κB in RAW264.7 cells. In mice, N-acetyl-l-cysteine or PDTC prevented the paclitaxel-induced allodynia. Co-culture of neuron-like NG108-15 cells or stimulation with their conditioned medium promoted paclitaxel-induced HMGB1 release from RAW264.7 cells. Our data indicate that HMGB1 released from macrophages through the ROS/p38MAPK/NF-κB/HAT pathway participates in the paclitaxel-induced peripheral neuropathy in mice, and unveils an emerging therapeutic avenue targeting a neuroimmune crosstalk in CIPN.


Subject(s)
HMGB1 Protein/metabolism , Macrophages/drug effects , Macrophages/metabolism , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/immunology , Acetylcysteine/pharmacology , Animals , Antibodies/pharmacology , Cells, Cultured , Clodronic Acid/pharmacology , Coculture Techniques , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/complications , Hyperalgesia/prevention & control , Imidazoles/pharmacology , Male , Membrane Proteins/metabolism , Mice , Minocycline/pharmacology , Neurons/metabolism , Paclitaxel/antagonists & inhibitors , Peripheral Nervous System Diseases/complications , Phosphoproteins/metabolism , Phosphorylation/drug effects , Proline/analogs & derivatives , Proline/pharmacology , Pyridines/pharmacology , Pyruvates/pharmacology , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/metabolism , Receptors, CXCR4 , Recombinant Proteins/metabolism , Sciatic Nerve/drug effects , Thiocarbamates/pharmacology , Thrombomodulin/metabolism , Up-Regulation/drug effects , p300-CBP Transcription Factors/metabolism
5.
Cell Death Dis ; 8(6): e2848, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28569790

ABSTRACT

Drug resistance is a major problem in cancer therapy. A growing body of evidence demonstrates that the tumor microenvironment, including cancer-associated fibroblasts (CAFs), can modulate drug sensitivity in tumor cells. We examined the effect of primary human CAFs on p53 induction and cell viability in prostate cancer cells on treatment with chemotherapeutic drugs. Co-culture with prostate CAFs or CAF-conditioned medium attenuated DNA damage and the p53 response to chemotherapeutic drugs and enhanced prostate cancer cell survival. CAF-conditioned medium inhibited the accumulation of doxorubicin, but not taxol, in prostate cancer cells in a manner that was associated with increased cancer cell glutathione levels. A low molecular weight fraction (<3 kDa) of CAF-conditioned medium had the same effect. CAF-conditioned medium also inhibited induction of reactive oxygen species (ROS) in both doxorubicin- and taxol-treated cancer cells. Our findings suggest that CAFs can enhance drug resistance in cancer cells by inhibiting drug accumulation and counteracting drug-induced oxidative stress. This protective mechanism may represent a novel therapeutic target in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer-Associated Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic , Glutathione/agonists , Prostatic Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , DNA Damage , Doxorubicin/antagonists & inhibitors , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Glutathione/metabolism , Humans , Male , Paclitaxel/antagonists & inhibitors , Paclitaxel/pharmacology , Primary Cell Culture , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Microenvironment/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
6.
Biol Pharm Bull ; 40(4): 473-478, 2017.
Article in English | MEDLINE | ID: mdl-28381802

ABSTRACT

Paclitaxel is a chemotherapeutic agent that causes peripheral neuropathy as its major dose-limiting side effect. However, the peripheral neuropathy is difficult to manage. A study we recently conducted showed that repetitive administration of aucubin as a prophylactic inhibits paclitaxel-induced mechanical allodynia. However, the mechanisms underlying the anti-allodynic activity of aucubin, which is a major component of Plantaginis Semen, was unclear. In addition to mechanical allodynia, aucubin inhibited spontaneous and mechanical stimuli-induced firing in spinal dorsal horn neurons; however, catalpol, a metabolite of aucubin, did not show these effects. Furthermore, paclitaxel induced the expression of CCAAT/enhancer-binding protein homologous protein, a marker of endoplasmic reticulum (ER) stress, in the sciatic nerve and a Schwann cell line (LY-PPB6 cells); however, this effect was inhibited by aucubin. These results suggest that aucubin inhibits paclitaxel-induced mechanical allodynia through the inhibition of ER stress in peripheral Schwann cells.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Hyperalgesia/prevention & control , Iridoid Glucosides/administration & dosage , Paclitaxel/toxicity , Pre-Exposure Prophylaxis/methods , Schwann Cells/drug effects , Animals , Antineoplastic Agents, Phytogenic/toxicity , Cell Line , Endoplasmic Reticulum Stress/physiology , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Male , Mice , Mice, Inbred C57BL , Paclitaxel/antagonists & inhibitors , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Rats , Schwann Cells/metabolism
7.
Pharmazie ; 71(12): 727-732, 2016 12 01.
Article in English | MEDLINE | ID: mdl-29442003

ABSTRACT

Methylsulfinylmethane (dimethyl sulfoxide; DMSO) is widely used in clinical treatment and bioresearch. Moreover, there is bioconversion between methylsulfanylmethane (dimethyl sulfide; DMS), DMSO, and methylsulfonylmethane (DMSO2) in mammalian metabolism. Due to the real-time detection limits for volatile compounds, most research has focused on DMSO2 as a stable byproduct of DMSO. Therefore, details about the production of DMS as a byproduct of DMSO metabolism remain to be elucidated. Here, we report the characterization of trace-level volatile organic compounds (VOCs) produced following DMSO treatment of cultured human cells using an ultrasensitive vacuum ultraviolet photoionization mass spectrometer (VUV-PIMS). Using this approach, 24 h after DMSO treatment we detected 16.9 and 21 parts per billion by volume (ppbv) DMS in the atmosphere above the cells (headspace) within HeLa and 293T tissue culture flasks, respectively. When simultaneously exposed to 50 nM paclitaxel (PTX), 17.6 and 22.3 ppbv DMS were detected in the headspace of HeLa and 293T culture flasks, respectively. Nevertheless, at doses of PTX more or less than 50 nM, the detectable levels of DMS were reduced to as low as 8.4 ppbv. Our experimental results demonstrate that by co-administering 5 to 10 nM PTX with DMSO, it is possible to moderate the production of DMS considerably. However, at higher doses of PTX, increased apoptosis was observed that likely contributed to higher DMS production by cells.


Subject(s)
Cell Survival/drug effects , Dimethyl Sulfoxide/pharmacology , Protective Agents/pharmacology , Volatile Organic Compounds/metabolism , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/toxicity , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans , Paclitaxel/antagonists & inhibitors , Paclitaxel/toxicity , Sulfones
8.
J Neurosci ; 35(39): 13487-500, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424893

ABSTRACT

Peripheral neuropathy is dose limiting in paclitaxel cancer chemotherapy and can result in both acute pain during treatment and chronic persistent pain in cancer survivors. The hypothesis tested was that paclitaxel produces these adverse effects at least in part by sensitizing transient receptor potential vanilloid subtype 1 (TRPV1) through Toll-like receptor 4 (TLR4) signaling. The data show that paclitaxel-induced behavioral hypersensitivity is prevented and reversed by spinal administration of a TRPV1 antagonist. The number of TRPV1(+) neurons is increased in the dorsal root ganglia (DRG) in paclitaxel-treated rats and is colocalized with TLR4 in rat and human DRG neurons. Cotreatment of rats with lipopolysaccharide from the photosynthetic bacterium Rhodobacter sphaeroides (LPS-RS), a TLR4 inhibitor, prevents the increase in numbers of TRPV1(+) neurons by paclitaxel treatment. Perfusion of paclitaxel or the archetypal TLR4 agonist LPS activated both rat DRG and spinal neurons directly and produced acute sensitization of TRPV1 in both groups of cells via a TLR4-mediated mechanism. Paclitaxel and LPS sensitize TRPV1 in HEK293 cells stably expressing human TLR4 and transiently expressing human TRPV1. These physiological effects also are prevented by LPS-RS. Finally, paclitaxel activates and sensitizes TRPV1 responses directly in dissociated human DRG neurons. In summary, TLR4 was activated by paclitaxel and led to sensitization of TRPV1. This mechanism could contribute to paclitaxel-induced acute pain and chronic painful neuropathy. Significance statement: In this original work, it is shown for the first time that paclitaxel activates peripheral sensory and spinal neurons directly and sensitizes these cells to transient receptor potential vanilloid subtype 1 (TRPV1)-mediated capsaicin responses via Toll-like receptor 4 (TLR4) in multiple species. A direct functional interaction between TLR4 and TRPV1 is shown in rat and human dorsal root ganglion neurons, TLR4/TRPV1-coexpressing HEK293 cells, and in both rat and mouse spinal cord slices. Moreover, this is the first study to show that this interaction plays an important role in the generation of behavioral hypersensitivity in paclitaxel-related neuropathy. The key translational implications are that TLR4 and TRPV1 antagonists may be useful in the prevention and treatment of chemotherapy-induced peripheral neuropathy in humans.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/pharmacology , Sensory Receptor Cells/drug effects , TRPV Cation Channels/antagonists & inhibitors , Toll-Like Receptor 4/drug effects , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Calcium/metabolism , Excitatory Postsynaptic Potentials/drug effects , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Paclitaxel/antagonists & inhibitors , Pain Measurement/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord/drug effects , Toll-Like Receptor 4/antagonists & inhibitors
9.
Br J Pharmacol ; 171(3): 636-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24117398

ABSTRACT

BACKGROUND AND PURPOSE: Paclitaxel (PAC) is associated with chemotherapy-induced neuropathic pain (CIPN) that can lead to the cessation of treatment in cancer patients even in the absence of alternate therapies. We previously reported that chronic administration of the non-psychoactive cannabinoid cannabidiol (CBD) prevents PAC-induced mechanical and thermal sensitivity in mice. Hence, we sought to determine receptor mechanisms by which CBD inhibits CIPN and whether CBD negatively effects nervous system function or chemotherapy efficacy. EXPERIMENTAL APPROACH: The ability of acute CBD pretreatment to prevent PAC-induced mechanical sensitivity was assessed, as was the effect of CBD on place conditioning and on an operant-conditioned learning and memory task. The potential interaction of CBD and PAC on breast cancer cell viability was determined using the MTT assay. KEY RESULTS: PAC-induced mechanical sensitivity was prevented by administration of CBD (2.5 - 10 mg·kg⁻¹) in female C57Bl/6 mice. This effect was reversed by co-administration of the 5-HT(1A) antagonist WAY 100635, but not the CB1 antagonist SR141716 or the CB2 antagonist SR144528. CBD produced no conditioned rewarding effects and did not affect conditioned learning and memory. Also, CBD + PAC combinations produce additive to synergistic inhibition of breast cancer cell viability. CONCLUSIONS AND IMPLICATIONS: Our data suggest that CBD is protective against PAC-induced neurotoxicity mediated in part by the 5-HT(1A) receptor system. Furthermore, CBD treatment was devoid of conditioned rewarding effects or cognitive impairment and did not attenuate PAC-induced inhibition of breast cancer cell viability. Hence, adjunct treatment with CBD during PAC chemotherapy may be safe and effective in the prevention or attenuation of CIPN.


Subject(s)
Cannabidiol/therapeutic use , Neuralgia/prevention & control , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Paclitaxel/antagonists & inhibitors , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/agonists , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Breast Neoplasms/drug therapy , Cannabidiol/adverse effects , Cannabidiol/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Conditioning, Operant/drug effects , Drug Synergism , Female , Humans , Memory/drug effects , Mice , Mice, Inbred C57BL , Neuralgia/chemically induced , Neuralgia/metabolism , Neuralgia/physiopathology , Neurons/metabolism , Neuroprotective Agents/adverse effects , Neuroprotective Agents/antagonists & inhibitors , Paclitaxel/adverse effects , Paclitaxel/agonists , Paclitaxel/pharmacology , Receptor, Serotonin, 5-HT1A/chemistry , Serotonin 5-HT1 Receptor Agonists/adverse effects , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Antagonists/pharmacology
10.
Ann Neurol ; 74(6): 893-904, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23955554

ABSTRACT

OBJECTIVE: Peripheral neurotoxicity is a major dose-limiting side effect of many chemotherapeutic drugs. Currently there are no effective disease-modifying therapies for chemotherapy-induced peripheral neuropathies, but these side effects of chemotherapy are potentially ideal targets for development of neuroprotective therapies, because candidate drugs can be co- or preadministered before the injury to peripheral axons takes place. METHODS: We used a phenotypic drug screening approach to identify ethoxyquin as a potential neuroprotective drug and carried out additional biochemical experiments to identify its mechanism of action. RESULTS: We validated the screening results with ethoxyquin and its derivatives and showed that they prevented paclitaxel-induced peripheral neuropathy without blocking paclitaxel's ability to kill tumor cells. Furthermore, we demonstrated that ethoxyquin acts by modulating the chaperone activity of heat shock protein 90 (Hsp90) and blocking the binding of 2 of its client proteins, ataxin-2 and Sf3b2. Ethoxyquin-induced reduction in levels of both of these proteins resulted in prevention of axonal degeneration caused by paclitaxel. INTERPRETATION: Ethoxyquin and its novel derivatives as well as other classes of small molecules that act as Hsp90 modulators may offer a new opportunity for development of drugs to prevent chemotherapy-induced axonal degeneration.


Subject(s)
Drug Evaluation, Preclinical/methods , Ethoxyquin/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Axons/drug effects , Cell Line , Male , Mice , Nerve Degeneration/chemically induced , Neurons/drug effects , Paclitaxel/adverse effects , Paclitaxel/antagonists & inhibitors
11.
Neuroscience ; 244: 77-89, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23583762

ABSTRACT

Besides its prominent role in angiogenesis, the vascular endothelial growth factor (VEGF) also exerts important protective effects on neurons. In particular, mice expressing reduced levels of VEGF suffer from late-onset motor neuron degeneration, whereas VEGF delivery significantly delays motor neuron death in ALS mouse models, at least partly through neuroprotective effects. Additionally, VEGF protects dorsal root ganglion (DRG) neurons against paclitaxel-induced neurotoxicity. Here, we demonstrate that VEGF also protects DRG neurons against hyperglycemia-induced neuronal stress as a model of diabetes-induced peripheral neuropathy. Specifically, VEGF decreased expression of the stress-related gene activating transcription factor 3 (ATF3) in DRG neurons isolated from streptozotocin-induced diabetic mice (ex vivo) and in isolated DRG neurons exposed to high glucose concentrations (in vitro). In vivo, local VEGF application also protected against paclitaxel- and diabetes-induced neuropathies without causing side effects. A small synthetic VEGF mimicking pentadecapeptide (QK) exerted similar effects on DRG cultures: the peptide reduced ATF3 expression in vitro and ex vivo in paclitaxel- and hyperglycemia-induced models of neuropathy to a similar extent as the full-length recombinant VEGF protein. By using transgenic mice selectively overexpressing the VEGF receptor 2 in postnatal neurons, these neuroprotective effects were shown to be mediated through VEGF receptor 2. Overall, these results underscore the potential of VEGF and VEGF-derived peptides for the treatment of peripheral neuropathies.


Subject(s)
Activating Transcription Factor 3/metabolism , Drug Evaluation, Preclinical , Ganglia, Spinal/drug effects , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Peripheral Nervous System Diseases/drug therapy , Vascular Endothelial Growth Factor A/pharmacology , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Ganglia, Spinal/metabolism , Glucose/antagonists & inhibitors , Glucose/toxicity , Male , Mice , Mice, Transgenic , Neuroprotective Agents/therapeutic use , Paclitaxel/antagonists & inhibitors , Paclitaxel/toxicity , Peptide Fragments/therapeutic use , Rats , Vascular Endothelial Growth Factor A/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/genetics
12.
Anesth Analg ; 114(5): 1104-20, 2012 May.
Article in English | MEDLINE | ID: mdl-22392969

ABSTRACT

BACKGROUND: Peripheral neuropathy is a major dose-limiting toxicity of chemotherapy, especially after multiple courses of paclitaxel. The development of paclitaxel-induced neuropathy is associated with the activation of microglia followed by the activation and proliferation of astrocytes, and the expression and release of proinflammatory cytokines in the spinal dorsal horn. Cannabinoid type 2 (CB(2)) receptors are expressed in the microglia in neurodegenerative disease models. METHODS: To explore the potential of CB(2) agonists for preventing paclitaxel-induced neuropathy, we designed and synthesized a novel CB(2)-selective agonist, namely, MDA7. The effect of MDA7 in preventing paclitaxel-induced allodynia was assessed in rats and in CB(2)(+/+) and CB(2)(-/-) mice. We hypothesized that the CB(2) receptor functions in a negative-feedback loop and that early MDA7 administration can blunt the neuroinflammatory response to paclitaxel and prevent mechanical allodynia through interference with specific signaling pathways. RESULTS: We found that MDA7 prevents paclitaxel-induced mechanical allodynia in rats and mice in a dose- and time-dependent manner without compromising paclitaxel's antineoplastic effect. MDA7's neuroprotective effect was absent in CB(2)(-/-) mice and was blocked by CB(2) antagonists, suggesting that MDA7's action directly involves CB(2) receptor activation. MDA7 treatment was found to interfere with early events in the paclitaxel-induced neuroinflammatory response as evidenced by relatively reduced toll-like receptor and CB(2) expression in the lumbar spinal cord, reduced levels of extracellular signal-regulated kinase 1/2 activity, reduced numbers of activated microglia and astrocytes, and reduced secretion of proinflammatory mediators in vivo and in in vitro models. CONCLUSIONS: Our findings suggest an innovative therapeutic approach to prevent chemotherapy-induced neuropathy and may permit more aggressive use of active chemotherapeutic regimens with reduced long-term sequelae.


Subject(s)
Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/toxicity , Benzofurans/pharmacology , Neuroprotective Agents , Paclitaxel/antagonists & inhibitors , Paclitaxel/toxicity , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control , Piperidines/pharmacology , Receptor, Cannabinoid, CB2/agonists , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Benzofurans/pharmacokinetics , Blotting, Western , CD11b Antigen/metabolism , Cricetinae , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Humans , Hyperalgesia/chemically induced , Hyperalgesia/prevention & control , Image Processing, Computer-Assisted , Immunohistochemistry , Lipopolysaccharides , Male , Mice , Mice, Knockout , Microscopy, Confocal , Neuroglia/drug effects , Physical Stimulation , Piperidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Cannabinoid, CB2/biosynthesis , Receptor, Cannabinoid, CB2/genetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Toll-Like Receptor 2/biosynthesis
13.
Cancer Sci ; 102(11): 2038-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21801281

ABSTRACT

Patients with estrogen receptor (ER)-positive breast cancers have a better prognosis than those with ER-negative breast cancers, but often have low sensitivity to chemotherapy and a limited survival benefit. We have previously shown a combination effect of taxanes and fulvestrant and suggested that this treatment may be useful for ER-positive breast cancer. In this study, we evaluated the effects of combinations of hormone drugs and chemotherapeutic agents. In vitro, the effects of combinations of five chemotherapeutic agents (doxorubicin, paclitaxel, docetaxel, vinorelbine, and 5-fluorouracil) and three hormone drugs (fulvestrant, tamoxifen, and 4-hydroxytamoxifen) were examined in ER-positive breast cancer cell lines using CalcuSyn software. Changes in chemoresistant factors such as Bcl2, multidrug resistance-associated protein 1, and microtubule-associated protein tau were also examined after exposure of the cells to hormone drugs. In vivo, tumor sizes in mice were evaluated after treatment with docetaxel or doxorubicin alone, fulvestrant alone, and combinations of these agents. Combination treatment with fulvestrant and all five chemotherapeutic agents in vitro showed synergistic effects. In contrast, tamoxifen showed an antagonistic effect with all the chemotherapeutic agents. 4-Hydroxytamoxifen showed an antagonistic effect with doxorubicin and 5-fluorouracil, but a synergistic effect with taxanes and vinorelbine. Regarding chemoresistant factors, Bcl2 and microtubule-associated protein tau were downregulated by fulvestrant. In vivo, a combination of fulvestrant and docetaxel had a synergistic effect on tumor growth, but fulvestrant and doxorubicin did not show this effect. In conclusion, fulvestrant showed good compatibility with all the evaluated chemotherapeutic agents, and especially with docetaxel, in vitro and in vivo.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Estradiol/analogs & derivatives , Estrogens , Neoplasms, Hormone-Dependent/pathology , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Docetaxel , Doxorubicin/administration & dosage , Doxorubicin/antagonists & inhibitors , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Estradiol/administration & dosage , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Female , Fluorouracil/administration & dosage , Fluorouracil/antagonists & inhibitors , Fluorouracil/pharmacology , Fulvestrant , Humans , Mice , Mice, Nude , Neoplasm Proteins/biosynthesis , Paclitaxel/administration & dosage , Paclitaxel/antagonists & inhibitors , Paclitaxel/pharmacology , Random Allocation , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Taxoids/administration & dosage , Taxoids/antagonists & inhibitors , Taxoids/pharmacology , Tumor Cells, Cultured/drug effects , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinblastine/antagonists & inhibitors , Vinblastine/pharmacology , Vinorelbine , Xenograft Model Antitumor Assays
14.
Anticancer Drugs ; 22(1): 58-78, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20890178

ABSTRACT

The aim was to determine the potential of the allosteric mammalian target of rapamycin inhibitor, everolimus, to act in combination with cytotoxic anticancer compounds in vitro and in vivo. A concomitant combination in vitro showed no evidence of antagonism, but enhanced the antiproliferative effects (additive to synergistic) with cisplatin, doxorubicin, 5-fluorouracil, gemcitabine, paclitaxel, and patupilone. Everolimus (1-5 mg/kg/d orally) was evaluated for antitumor activity in vivo alone or in combination with suboptimal cytotoxic doses using athymic nude mice bearing subcutaneous human H-596 lung, KB-31 cervical, or HCT-116 colon tumor xenografts. Everolimus monotherapy was very well tolerated and caused inhibition of tumor growth, rather than regression, and this was associated with a dose-dependent decline in tumor pS6 levels, a key downstream protein of mammalian target of rapamycin. At the doses used, the cytotoxics inhibited tumor growth and caused tolerable body-weight loss. Concomitant combinations of cisplatin, doxorubicin, paclitaxel, or patupilone with everolimus produced cooperative antitumor effects, in some cases producing regressions without clinically significant increases in toxicity. In contrast, combinations with gemcitabine and 5-fluorouracil were less well tolerated. Alternative administration schedules were tested for cisplatin, gemcitabine, or paclitaxel combined with everolimus: these did not dramatically affect cisplatin or gemcitabine activity or tolerability but were antagonistic for paclitaxel. Everolimus showed promising maintenance activity after treatment with doxorubicin or paclitaxel ceased. Overall, the results confirm that everolimus is an effective, well-tolerated suppressor of experimental human tumor growth, and although it did not show strong potentiation of efficacy, antitumor activity in vivo was increased without marked increases in toxicity, supporting clinical use of everolimus as a partner for conventional cytotoxics.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Epothilones/administration & dosage , Everolimus , Female , Fluorouracil/administration & dosage , HCT116 Cells , Humans , Mice , Mice, Nude , Paclitaxel/administration & dosage , Paclitaxel/antagonists & inhibitors , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Xenograft Model Antitumor Assays , Gemcitabine
15.
Pain ; 152(2): 308-313, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21145656

ABSTRACT

Loss of intraepidermal nerve fibers (IENFs) has been speculated to play a critical role in the development of various neuropathies. In this study, the density of IENFs were studied over time during the induction of Taxol (Bristol-Myers Squibb, NY, USA)-induced chemoneuropathy and compared with the changes in IENFs in animals co-treated with Taxol plus the protective agent minocycline. Rats were injected (intraperitoneally) with 2mg/kg of Taxol every other day for four injections (day 1, 3, 5, and 7). Minocycline (25mg/kg) was given in a separate group of rats 24h prior to the first dose of Taxol and every day for the next 9days (day 0 through 9). Animals were tested for mechanical paw withdrawal thresholds prior to any drug administrations and again on day 7, 14, and 30. Immunohistochemistry using the pan-neuronal marker protein gene product 9.5 was performed on glabrous skin of the hind-paw foot pad to stain for IENFs also on day 7, 14, and 30. The results show that Taxol-treated animals developed mechanical sensitivity and corresponding IENF loss. Animals receiving minocycline plus Taxol showed no hyperalgesia or loss of IENFs. This study confirms, for the first time, that a loss of IENFs occurs as a neuropathy develops, and further shows a protection against both IENF loss and hyperalgesia with minocycline treatment. The progression of Taxol-induced mechanical hypersensitivity coincides with loss of intraepidermal nerve fibers, and the hyperalgesia and nerve fiber loss were prevented with minocycline treatment.


Subject(s)
Epidermis/innervation , Epidermis/pathology , Hyperalgesia/drug therapy , Minocycline/pharmacology , Nerve Fibers/pathology , Paclitaxel/toxicity , Animals , Disease Models, Animal , Epidermis/drug effects , Hyperalgesia/chemically induced , Hyperalgesia/pathology , Male , Minocycline/therapeutic use , Nerve Fibers/drug effects , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Paclitaxel/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
16.
Nutr Cancer ; 62(8): 1025-35, 2010.
Article in English | MEDLINE | ID: mdl-21058190

ABSTRACT

Quercetin is a flavonoid with anticancer properties. In this study, we examined the effects of quercetin on cell cycle, viability, and proliferation of cancer cells, either singly or in combination with the microtubule-targeting drugs taxol and nocodazole. Although quercetin induced cell death in a dose-dependent manner, 12.5-50 µM quercetin inhibited the activity of both taxol and nocodazole to induce G2/M arrest in various cell lines. Quercetin also partially restored drug-induced loss in viability of treated cells for up to 72 h. This antagonism of microtubule-targeting drugs was accompanied by a delay in cell cycle progression and inhibition of the buildup of cyclin-B1 at the microtubule organizing center of treated cells. However, quercetin did not inhibit the microtubule targeting of taxol or nocodazole. Despite the short-term protection of cells by quercetin, colony formation and clonogenicity of HCT116 cells were still suppressed by quercetin or quercetin-taxol combination. The status of cell adherence to growth matrix was critical in determining the sensitivity of HCT116 cells to quercetin. We conclude that although long-term exposure of cancer cells to quercetin may prevent cell proliferation and survival, the interference of quercetin with cell cycle progression diminishes the efficacy of microtubule-targeting drugs to arrest cells at G2/M.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Cell Cycle/drug effects , Neoplasms/drug therapy , Nocodazole/antagonists & inhibitors , Paclitaxel/antagonists & inhibitors , Quercetin/metabolism , Tubulin Modulators/antagonists & inhibitors , Cell Adhesion , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin B1/metabolism , Food-Drug Interactions , G2 Phase/drug effects , Humans , Microtubules/drug effects , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control , Nocodazole/pharmacology , Osmolar Concentration , Paclitaxel/pharmacology , Time Factors , Tubulin/metabolism , Tubulin Modulators/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Mol Nutr Food Res ; 54(11): 1574-84, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20521268

ABSTRACT

Resveratrol, which is highly concentrated in the skin of grapes and is abundant in red wine, has been demonstrated to account for several beneficial properties, including antioxidant, anticoagulant, anti-inflammatory and anticancer effects. Taxol is a microtubule-stabilizing drug that has been extensively used as effective chemotherapeutic agents in the treatment of solid tumors. Here, we investigated whether the combination of the two compounds would yield increased antitumor efficacy in human cancer cells. Unexpectedly, resveratrol effectively prevented tumor cell death induced by taxol in 5637 bladder cancer cells. This pronounced antagonistic function of resveratrol against taxol was associated with changes in multiple signal transduction pathways, but not with tubulin polymerization. Importantly, cell cycle analysis showed that resveratrol prevented the cells from entering into mitosis, the phase in which taxol exerts its action. Furthermore, resveratrol blocked the cytotoxic effects of vinblastine but not cisplatin in 5637 cells. Interestingly, resveratrol pre-treatment followed by taxol resulted in synergistic cytotoxicity. Finally, we extended our studies to various human cancer cell lines. Taken together, our results indicate that resveratrol may have the potential to negate the therapeutic efficacy of taxol and suggest that consumption of resveratrol-related products may be contraindicated during cancer therapy with taxol.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Cell Cycle/drug effects , Paclitaxel/antagonists & inhibitors , Stilbenes/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Humans , Mitosis/drug effects , Polymerization , Resveratrol , Signal Transduction , Vinblastine/antagonists & inhibitors
18.
Eur J Cancer ; 46(10): 1882-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20223651

ABSTRACT

It was reported recently that resveratrol could sensitise a number of cancer cell lines to the anticancer actions of several other cancer drugs, including paclitaxel. In the present study, we further investigated whether resveratrol could sensitise human breast cancer cells to paclitaxel-induced cell death. Unexpectedly, we found that resveratrol strongly diminished the susceptibility of MDA-MB-435s, MDA-MB-231 and SKBR-3 cells to paclitaxel-induced cell death in culture, although this effect was not observed in MCF-7 cells. Using MDA-MB-435s cells as a representative model, a similar observation was made in athymic nude mice. Mechanistically, the modulating effect of resveratrol was partially attributable to its inhibition of paclitaxel-induced G(2)/M cell cycle arrest, together with an accumulation of cells in the S-phase. In addition, resveratrol could suppress paclitaxel-induced accumulation of reactive oxygen species (ROS) and subsequently the inactivation of anti-apoptotic Bcl-2 family proteins. These observations suggest that the strategy of concomitant use of resveratrol with paclitaxel is detrimental in certain types of human cancers. Given the widespread use of resveratrol among cancer patients, this study calls for more preclinical and clinical testing of the potential benefits and harms of using resveratrol as a dietary adjuvant in cancer patients.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Breast Neoplasms/drug therapy , Paclitaxel/antagonists & inhibitors , Stilbenes/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Resveratrol , Transplantation, Heterologous , bcl-X Protein/metabolism
19.
Clin Cancer Res ; 16(2): 600-9, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068074

ABSTRACT

PURPOSE: Ovarian cancer recurrence with the development of paclitaxel resistance is an obstacle to long-term survival. We showed that secretory leukocyte protease inhibitor (SLPI) is a survival factor for ovarian cancer. We hypothesize that SLPI may antagonize paclitaxel injury. EXPERIMENTAL DESIGN: Differential SLPI induction in response to paclitaxel and in response to stable forced expression of SLPI was shown in A2780-1A9 cells and their paclitaxel-resistant sublines, PTX10 and PTX22, and confirmed with HEY-A8 cells. SLPI-mediated survival was reduced by the MAP/extracellular signal-regulated kinase (ERK) kinase inhibitor, U0126, and a humanized neutralizing monoclonal anti-SLPI antibody, CR012. OVCAR3 xenographs tested the role of CR012 in vivo. RESULTS: SLPI expression was lower in A2780-1A9 ovarian cancer cells than in PTX10 and PTX22, and SLPI was induced by paclitaxel exposure. Stable SLPI expression yielded a proliferation advantage (P = 0.01); expression of and response to SLPI in OVCAR3 cells were abrogated by exposure to CR012. SLPI reduced the paclitaxel susceptibility of 1A9 and HEY-A8 cells (P

Subject(s)
Carcinoma/pathology , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/pathology , Paclitaxel/antagonists & inhibitors , Secretory Leukocyte Peptidase Inhibitor/physiology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/drug therapy , Carcinoma/genetics , Carcinoma/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Drug Antagonism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, SCID , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Secretory Leukocyte Peptidase Inhibitor/genetics , Secretory Leukocyte Peptidase Inhibitor/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Anticancer Res ; 29(10): 3995-4003, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19846942

ABSTRACT

BACKGROUND: Tetracyclines such as doxycycline are reported to possess cytotoxic activity against mammalian tumor cells, but the mechanism of their effects on cell proliferation remains unclear. MATERIALS AND METHODS: The antitumor effect of doxycycline was investigated in human pancreatic cancer cell line, PANC-1. We also investigated the effect of doxycycline on expression of a potent proangiogenic factor, interleukin (IL)-8. RESULTS: In excess of 20 microg/ml, cytotoxic effects of doxycycline were accompanied by G(1)-S cell cycle arrest and DNA fragmentation in PANC-1 cells. Doxycycline consistently activated transcription of p53, p21 and Fas/FasL-cascade-related genes, while reducing the expression of Bcl-xL and Mcl-1. Doxycycline (5 microg/ml) below the cytotoxic level suppressed endogenous and paclitaxel-induced IL-8 expression. In the mouse xenograft model, doxycycline treatment was shown to suppress tumor growth by 80%. CONCLUSION: These data suggest that doxycycline exerts its antitumor effect by activating proapoptotic genes, inhibiting IL-8 expression, and suppressing antiapoptotic genes.


Subject(s)
Adenocarcinoma/drug therapy , Apoptosis/drug effects , Doxycycline/pharmacology , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis/genetics , Cell Growth Processes/drug effects , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Down-Regulation/drug effects , Drug Interactions , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-8/biosynthesis , Mice , Paclitaxel/antagonists & inhibitors , Paclitaxel/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , S Phase/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Protein p53/biosynthesis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...