Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Medicine (Baltimore) ; 99(25): e20809, 2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32569228

ABSTRACT

RATIONALE: Pyrotinib is a novel dual pan-ErbB receptor tyrosine kinase inhibitor, approved for the treatment of human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC). However, there was still limited information regarding specific effect of pyrotinib on HER2-positive MBC patients with phosphoinositol-3 kinase mutation. PATIENT CONCERNS: A 63-year-old woman accidentally discovered a left breast lesion. The breast cancer was diagnosed by biopsy of breast lesion and postoperative pathological examination in March, 2017. The patient was presented with HER2-positive (3+), invasive carcinoma of the left breast with lymph nodes and lung nodules metastasis, and the clinical stage was T4N2M1. However, the lesion continued to aggressive disease progression with the treatment of trastuzumab plus multiple chemotherapy regimens and traditional Chinese medicine. DIAGNOSES: The woman was diagnosed with invasive carcinoma of the left breast and lymph nodes and lung nodules metastasis. INTERVENTIONS: The patient received 6 cycles of pyrotinib in combination with capecitabine regularly. OUTCOMES: Progression free survival was more than 6 months, and the patient's efficacy evaluation was partial remission. LESSONS: Our clinical observations demonstrated that pyrotinib may be an effective treatment for patients with HER2-positive MBC.


Subject(s)
Acrylamides/therapeutic use , Aminoquinolines/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Carcinoma, Ductal, Breast/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Acrylamides/administration & dosage , Aminoquinolines/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Capecitabine/administration & dosage , Capecitabine/therapeutic use , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Female , Humans , Middle Aged
2.
Toxicol Appl Pharmacol ; 379: 114662, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31301315

ABSTRACT

Although osimertinib, an EGFR tyrosine kinase inhibitor, has become the standard therapy for treating non-small cell lung cancer (NSCLC) patients with EGFR-activating mutation, upregulation of MCL-1 induces acquired resistance to osimertinib. Bufalin, a natural digoxin-like ingredient isolated from a traditional Chinese medicine Chan Su, has been shown to downregulate MCL-1 in NSCLC cells. However, whether bufalin reverses this acquired resistance to osimertinib in NSCLC cells remains unclear. In this study, bufalin reduced cell viability and promoted apoptosis in osimertinib-resistant cells. Moreover, co-treatment with bufalin and osimertinib restored the sensitivity of osimertinib-resistant cells to osimertinib-induced growth regression and apoptosis in vitro and in vivo. Mechanistically, MEK/ERK-dependent MCL-1 phosphorylation and Ku70-mediated MCL-1 overexpression confer osimertinib resistance in EGFR-mutant NSCLC cells. In osimertinib-resistant cells, bufalin modulates Ku70-mediated MCL-1 degradation, but not MEK/ERK/MCL-1 signaling. In conclusion, our study suggests that bufalin eliminates resistance to osimertinib by inhibiting Ku70-mediated MCL-1 overexpression, indicating that a combination of osimertinib and bufalin could be an effective additional treatment to overcome acquired resistance to osimertinib in NSCLC cells.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Bufanolides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Genes, erbB-1/genetics , Lung Neoplasms/drug therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Bufanolides/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/drug effects , Fluorescent Antibody Technique , In Situ Nick-End Labeling , Lung Neoplasms/genetics , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation
4.
Nat Commun ; 9(1): 4030, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30279412

ABSTRACT

Asthma accounts for 380,000 deaths a year. Carotid body denervation has been shown to have a profound effect on airway hyper-responsiveness in animal models but a mechanistic explanation is lacking. Here we demonstrate, using a rat model of asthma (OVA-sensitized), that carotid body activation during airborne allergic provocation is caused by systemic release of lysophosphatidic acid (LPA). Carotid body activation by LPA involves TRPV1 and LPA-specific receptors, and induces parasympathetic (vagal) activity. We demonstrate that this activation is sufficient to cause acute bronchoconstriction. Moreover, we show that prophylactic administration of TRPV1 (AMG9810) and LPA (BrP-LPA) receptor antagonists prevents bradykinin-induced asthmatic bronchoconstriction and, if administered following allergen exposure, reduces the associated respiratory distress. Our discovery provides mechanistic insight into the critical roles of carotid body LPA receptors in allergen-induced respiratory distress and suggests alternate treatment options for asthma.


Subject(s)
Acrylamides/therapeutic use , Asthma/prevention & control , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carotid Body/metabolism , Lysophospholipids/therapeutic use , Receptors, Lysophosphatidic Acid/metabolism , TRPV Cation Channels/metabolism , Acrylamides/pharmacology , Animals , Asthma/etiology , Asthma/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Lysophospholipids/pharmacology , Male , Rats, Inbred BN , Rats, Sprague-Dawley , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , TRPV Cation Channels/antagonists & inhibitors
5.
Epilepsy Res ; 145: 153-159, 2018 09.
Article in English | MEDLINE | ID: mdl-30007240

ABSTRACT

OBJECTIVE: Acetaminophen is one of the most commonly used analgesic and antipyretic drugs. It has been reported that acetaminophen has anticonvulsant effects in several animal models of seizure. An active metabolite of acetaminophen, AM404, inhibits the uptake of the endocannabinoid anandamide. However, the mechanism of the anticonvulsant effect of acetaminophen is unknown. METHODS: This study was performed to examine whether or not acetaminophen can protect against pentylenetetrazol-induced kindling in mice and to investigate the precise mechanisms of the anticonvulsant effect of acetaminophen using the fully kindled mouse models. RESULTS: Repeated administration of acetaminophen significantly delayed the progression of seizure severity induced by pentylenetetrazol. Additionally, acetaminophen showed a dose-dependent anticonvulsant activity against fully pentylenetetrazol-kindled seizures. AM404 also exhibited a dose-dependent anticonvulsant activity in fully kindled animals. The anticonvulsant activity of acetaminophen was antagonized by capsazepine and AMG9810, two transient receptor potential vanilloid-1 (TRPV1) antagonists. However, the transient receptor potential ankyrin 1 (TRPA1) antagonist HC030031 and CB1 receptor antagonist AM251 had no effect. CONCLUSION: These findings suggest that acetaminophen has an anticonvulsant effect in pentylenetetrazol-kindled mouse models and TRPV1 mediates the anticonvulsant action.


Subject(s)
Acetaminophen/therapeutic use , Anticonvulsants/therapeutic use , Seizures/drug therapy , TRPV Cation Channels/metabolism , Acetanilides/therapeutic use , Acrylamides/therapeutic use , Animals , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Kindling, Neurologic/drug effects , Male , Mice , Mice, Inbred ICR , Pentylenetetrazole/toxicity , Piperidines/therapeutic use , Purines/therapeutic use , Pyrazoles/therapeutic use , Seizures/chemically induced , TRPV Cation Channels/antagonists & inhibitors , Time Factors
6.
Circulation ; 137(21): 2256-2273, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29217642

ABSTRACT

BACKGROUND: Myocardial metabolic impairment is a major feature in chronic heart failure. As the major coenzyme in fuel oxidation and oxidative phosphorylation and a substrate for enzymes signaling energy stress and oxidative stress response, nicotinamide adenine dinucleotide (NAD+) is emerging as a metabolic target in a number of diseases including heart failure. Little is known on the mechanisms regulating homeostasis of NAD+ in the failing heart. METHODS: To explore possible alterations of NAD+ homeostasis in the failing heart, we quantified the expression of NAD+ biosynthetic enzymes in the human failing heart and in the heart of a mouse model of dilated cardiomyopathy (DCM) triggered by Serum Response Factor transcription factor depletion in the heart (SRFHKO) or of cardiac hypertrophy triggered by transverse aorta constriction. We studied the impact of NAD+ precursor supplementation on cardiac function in both mouse models. RESULTS: We observed a 30% loss in levels of NAD+ in the murine failing heart of both DCM and transverse aorta constriction mice that was accompanied by a decrease in expression of the nicotinamide phosphoribosyltransferase enzyme that recycles the nicotinamide precursor, whereas the nicotinamide riboside kinase 2 (NMRK2) that phosphorylates the nicotinamide riboside precursor is increased, to a higher level in the DCM (40-fold) than in transverse aorta constriction (4-fold). This shift was also observed in human failing heart biopsies in comparison with nonfailing controls. We show that the Nmrk2 gene is an AMP-activated protein kinase and peroxisome proliferator-activated receptor α responsive gene that is activated by energy stress and NAD+ depletion in isolated rat cardiomyocytes. Nicotinamide riboside efficiently rescues NAD+ synthesis in response to FK866-mediated inhibition of nicotinamide phosphoribosyltransferase and stimulates glycolysis in cardiomyocytes. Accordingly, we show that nicotinamide riboside supplementation in food attenuates the development of heart failure in mice, more robustly in DCM, and partially after transverse aorta constriction, by stabilizing myocardial NAD+ levels in the failing heart. Nicotinamide riboside treatment also robustly increases the myocardial levels of 3 metabolites, nicotinic acid adenine dinucleotide, methylnicotinamide, and N1-methyl-4-pyridone-5-carboxamide, that can be used as validation biomarkers for the treatment. CONCLUSIONS: The data show that nicotinamide riboside, the most energy-efficient among NAD precursors, could be useful for treatment of heart failure, notably in the context of DCM, a disease with few therapeutic options.


Subject(s)
Cardiomyopathy, Dilated/drug therapy , Niacinamide/analogs & derivatives , AMP-Activated Protein Kinases/metabolism , Acrylamides/therapeutic use , Animals , Citric Acid/metabolism , Cytokines/genetics , Cytokines/metabolism , Dietary Supplements , Disease Models, Animal , Gene Expression Profiling , Heart Failure/prevention & control , Metabolome/drug effects , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NAD/metabolism , Niacinamide/therapeutic use , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , PPAR alpha/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Piperidines/therapeutic use , Pyridinium Compounds , Rats , Serum Response Factor/deficiency , Serum Response Factor/genetics
7.
World J Gastroenterol ; 23(33): 6065-6076, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28970722

ABSTRACT

AIM: To evaluate whether non-steroidal anti-inflammatory drugs (NSAIDs)-induced gastropathy is a clinically predictive model of referred visceral hypersensitivity. METHODS: Gastric ulcer pain was induced by the oral administration of indomethacin to male, CD1 mice (n = 10/group) and then assessed by measuring referred abdominal hypersensitivity to tactile application. A diverse range of pharmacological mechanisms contributing to the pain were subsequently investigated. These mechanisms included: transient receptor potential (TRP), sodium and acid-sensing ion channels (ASICs) as well as opioid receptors and guanylate cyclase C (GC-C). RESULTS: Results showed that two opioids and a GC-C agonist, morphine, asimadoline and linaclotide, respectively, the TRP antagonists, AMG9810 and HC-030031 and the sodium channel blocker, carbamazepine, elicited a dose- and/or time-dependent attenuation of referred visceral hypersensitivity, while the ASIC blocker, amiloride, was ineffective at all doses tested. CONCLUSION: Together, these findings implicate opioid receptors, GC-C, and sodium and TRP channel activation as possible mechanisms associated with visceral hypersensitivity. More importantly, these findings also validate NSAID-induced gastropathy as a sensitive and clinically predictive mouse model suitable for assessing novel molecules with potential pain-attenuating properties.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Hyperalgesia/pathology , Stomach Ulcer/complications , Visceral Pain/pathology , Acetanilides/therapeutic use , Acid Sensing Ion Channel Blockers/therapeutic use , Acid Sensing Ion Channels/metabolism , Acrylamides/therapeutic use , Amiloride/therapeutic use , Analgesics, Opioid/therapeutic use , Animals , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Mice , Morphine/therapeutic use , Pain Measurement/methods , Purines/therapeutic use , Random Allocation , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Stomach Ulcer/chemically induced , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism , Visceral Pain/etiology
8.
Clin Sci (Lond) ; 131(15): 1841-1857, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28533215

ABSTRACT

Inhibition of total histone deacetylases (HDACs) was phenomenally associated with the prevention of diabetic cardiomyopathy (DCM). However, which specific HDAC plays the key role in DCM remains unclear. The present study was designed to determine whether DCM can be prevented by specific inhibition of HDAC3 and to elucidate the mechanisms by which inhibition of HDAC3 prevents DCM. Type 1 diabetes OVE26 and age-matched wild-type (WT) mice were given the selective HDAC3 inhibitor RGFP966 or vehicle for 3 months. These mice were then killed immediately or 3 months later for cardiac function and pathological examination. HDAC3 activity was significantly increased in the heart of diabetic mice. Administration of RGFP966 significantly prevented DCM, as evidenced by improved diabetes-induced cardiac dysfunction, hypertrophy, and fibrosis, along with diminished cardiac oxidative stress, inflammation, and insulin resistance, not only in the mice killed immediately or 3 months later following the 3-month treatment. Furthermore, phosphorylated extracellular signal-regulated kinases (ERK) 1/2, a well-known initiator of cardiac hypertrophy, was significantly increased, while dual specificity phosphatase 5 (DUSP5), an ERK1/2 nuclear phosphatase, was substantially decreased in diabetic hearts. Both of these changes were prevented by RGFP966. Chromatin immunoprecipitation (ChIP) assay showed that HDAC3 inhibition elevated histone H3 acetylation on the DUSP5 gene promoter at both two time points. These findings suggest that diabetes-activated HDAC3 inhibits DUSP5 expression through deacetylating histone H3 on the primer region of DUSP5 gene, leading to the derepression of ERK1/2 and the initiation of DCM. The present study indicates the potential application of HDAC3 inhibitor for the prevention of DCM.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Diabetic Cardiomyopathies/prevention & control , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/drug effects , Acrylamides/therapeutic use , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/genetics , Drug Evaluation, Preclinical/methods , Dual-Specificity Phosphatases/metabolism , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histone Deacetylases/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice, Transgenic , Myocardium/enzymology , Oxidative Stress/drug effects , Phenylenediamines/therapeutic use , Receptor, Insulin/metabolism , Signal Transduction/drug effects
9.
Kidney Int ; 92(4): 922-933, 2017 10.
Article in English | MEDLINE | ID: mdl-28545714

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a common hereditary renal disease with no currently available targeted therapies. Based on the established connection between ß-catenin signaling and renal ciliopathies, and on data from our and other laboratories showing striking similarities of this disease and cancer, we evaluated the use of an orally bioavailable small molecule, KPT-9274 (a dual inhibitor of the protein kinase PAK4 and nicotinamide phosphoribosyl transferase), for treatment of ADPKD. Treatment of PKD-derived cells with this compound not only reduces PAK4 steady-state protein levels and regulates ß-catenin signaling, but also inhibits nicotinamide phosphoribosyl transferase, the rate-limiting enzyme in a key NAD salvage pathway. KPT-9274 can attenuate cellular proliferation and induce apoptosis associated with a decrease in active (phosphorylated) PAK4 and ß-catenin in several Pkd1-null murine cell lines, with a less pronounced effect on the corresponding phenotypically normal cells. Additionally, KPT-9274 shows inhibition of cystogenesis in an ex vivo model of cyclic AMP-induced cystogenesis as well as in the early stage Pkd1flox/flox:Pkhd1-Cre mouse model, the latter showing confirmation of specific anti-proliferative, apoptotic, and on-target effects. NAD biosynthetic attenuation by KPT-9274, while critical for highly proliferative cancer cells, does not appear to be important in the slower growing cystic epithelial cells during cystogenesis. KPT-9274 was not toxic in our ADPKD animal model or in other cancer models. Thus, this small molecule inhibitor could be evaluated in a clinical trial as a viable therapy of ADPKD.


Subject(s)
Acrylamides/pharmacology , Aminopyridines/pharmacology , Cytokines/metabolism , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Polycystic Kidney, Autosomal Dominant/drug therapy , p21-Activated Kinases/metabolism , Acrylamides/therapeutic use , Aminopyridines/therapeutic use , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Epithelial Cells , Female , Humans , Kidney/cytology , Male , Mice , Mice, Transgenic , Organ Culture Techniques , Phosphorylation , Polycystic Kidney, Autosomal Dominant/pathology , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , TRPP Cation Channels/genetics , beta Catenin/metabolism
10.
Pigment Cell Melanoma Res ; 29(1): 81-91, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26459162

ABSTRACT

Piper amides have a characteristic, unsaturated amide group and exhibit diverse biological activities, including proliferation and differentiation of melanocytes, although the molecular mechanisms underlying its antimelanogenesis effect remain unknown. We screened a selected chemical library of newly synthesized Piper amide derivatives and identified (E)-3-(4-(tert-butyl)phenyl)-N-(2,3-dihydrobenzo[b][1,4]dioxin-6-yl)acrylamide (NED-180) as one of the most potent compounds in suppressing melanogenesis. In murine melan-a melanocytes, NED-180 downregulated the expression of melanogenic regulatory proteins including tyrosinase, Tyrp1, Dct, and MITF. PI3K/Akt-dependent phosphorylation of GSK3ß by NED-180 decreases MITF phosphorylation and inhibits melanogenesis without any effects on cytotoxicity and proliferation. Furthermore, topical application of NED-180 significantly ameliorated UVB-induced skin hyperpigmentation in guinea pigs. Interestingly, data obtained using calcium imaging techniques suggested that NED-180 reduced the TPA-induced activation of TRPM1 (melastatin), which could explain the NED-180-induced inhibition of melanogenesis. All things taken together, NED-180 triggers activation of multiple pathways, such as PI3K and ERK, and inhibits TRPM1/TRPV1, leading to inhibition of melanogenesis.


Subject(s)
Acrylamides/therapeutic use , Amides/therapeutic use , Calcium/metabolism , Dioxanes/therapeutic use , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperpigmentation/drug therapy , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Piper/chemistry , TRPM Cation Channels/metabolism , Acrylamides/pharmacology , Amides/pharmacology , Animals , Dioxanes/pharmacology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Guinea Pigs , HEK293 Cells , Humans , Hyperpigmentation/enzymology , Intramolecular Oxidoreductases/metabolism , Melanins/metabolism , Mice , Models, Biological , Monophenol Monooxygenase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/drug effects
11.
Future Oncol ; 11(22): 3069-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26450446

ABSTRACT

Non-small-cell lung cancer (NSCLC) patients whose tumors have an EGFR-activating mutation develop acquired resistance after a median of 9-11 months from the beginning of treatment with erlotinib, gefitinib and afatinib. T790M mutation is the cause of this resistance in approximately 60% of cases. AZD9291 is an oral, irreversible, mutant-selective EGF receptor (EGFR) tyrosine kinase inhibitor (TKI) developed to have potency against EGFR mutations, including T790M mutation, while sparing wild-type EGFR. A Phase I trial of AZD9291 in EGFR-mutant NSCLC patients, demonstrated high activity, essentially among T790M-mutant tumors, with a manageable tolerability profile. Ongoing Phase III trials are evaluating AZD9291 in EGFR-mutant patients as first-line treatment compared with erlotinib and gefitinib; and as second-line treatment compared with chemotherapy after progression on EGFR TKI in T790M-mutant tumors. Better identification of T790M-mutant tumors post EGFR TKI relapse and mechanisms of resistance to AZD9291 are the future challenges. This article reviews the emerging data regarding AZD9291 in the treatment of patients with advanced NSCLC.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Acrylamides/chemistry , Acrylamides/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/diagnosis , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Disease Progression , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Humans , Lung Neoplasms/diagnosis , Neoplasm Recurrence, Local , Neoplasm Staging , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Retreatment , Treatment Outcome
12.
Anesth Analg ; 121(5): 1369-77, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26280585

ABSTRACT

BACKGROUND: Positive allosteric modulators (PAMs) facilitate endogenous neurotransmission and/or enhance the efficacy of agonists without directly acting on the orthosteric binding sites. In this regard, selective α7 nicotinic acetylcholine receptor type II PAMs display antinociceptive activity in rodent chronic inflammatory and neuropathic pain models. This study investigates whether 3-furan-2-yl-N-p-tolyl-acrylamide (PAM-2), a new putative α7-selective type II PAM, attenuates experimental inflammatory and neuropathic pains in mice. METHODS: We tested the activity of PAM-2 after intraperitoneal administration in 3 pain assays: the carrageenan-induced inflammatory pain, the complete Freund adjuvant-induced inflammatory pain, and the chronic constriction injury-induced neuropathic pain in mice. We also tested whether PAM-2 enhanced the effects of the selective α7 agonist choline in the mouse carrageenan test given intrathecally. Because the experience of pain has both sensory and affective dimensions, we also evaluated the effects of PAM-2 on acetic acid-induced aversion by using the conditioned place aversion test. RESULTS: We observed that systemic administration of PAM-2 significantly reversed mechanical allodynia and thermal hyperalgesia in inflammatory and neuropathic pain models in a dose- and time-dependent manner without motor impairment. In addition, by attenuating the paw edema in inflammatory models, PAM-2 showed antiinflammatory properties. The antinociceptive effect of PAM-2 was inhibited by the selective competitive antagonist methyllycaconitine, indicating that the effect is mediated by α7 nicotinic acetylcholine receptors. Furthermore, PAM-2 enhanced the antiallodynic and antiinflammatory effects of choline, a selective α7 agonist, in the mouse carrageenan test. PAM-2 was also effective in reducing acetic acid-induced aversion in the conditioned place aversion assay. CONCLUSIONS: These findings suggest that the administration of PAM-2, a new α7-selective type II PAM, reduces the neuropathic and inflammatory pain sensory and affective behaviors in the mouse. Thus, this drug may have therapeutic applications in the treatment and management of chronic pain.


Subject(s)
Acrylamides/therapeutic use , Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Furans/therapeutic use , Pain/drug therapy , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/physiology , Acrylamides/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Furans/pharmacology , Male , Mice , Mice, Inbred ICR , Pain/pathology
13.
Biomaterials ; 63: 115-27, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26093792

ABSTRACT

To integrate biological imaging and multimodal therapies into one platform for enhanced anti-cancer efficacy, we have designed a novel core/shell structured nano-theranostic by conjugating photosensitive Au25(SR)18 - (SR refers to thiolate) clusters, pH/temperature-responsive polymer P(NIPAm-MAA), and anti-cancer drug (doxorubicin, DOX) onto the surface of mesoporous silica coated core-shell up-conversion nanoparticles (UCNPs). It is found that the photodynamic therapy (PDT) derived from the generated reactive oxygen species and the photothermal therapy (PTT) arising from the photothermal effect can be simultaneously triggered by a single 980 nm near infrared (NIR) light. Furthermore, the thermal effect can also stimulate the pH/temperature sensitive polymer in the cancer sites, thus realizing the targeted and controllable DOX release. The combined PDT, PTT and pH/temperature responsive chemo-therapy can markedly improve the therapeutic efficacy, which has been confirmed by both in intro and in vivo assays. Moreover, the doped rare earths endow the platform with dual-modal up-conversion luminescent (UCL) and computer tomography (CT) imaging properties, thus achieving the target of imaging-guided synergistic therapy under by a single NIR light.


Subject(s)
Acrylamides/chemistry , Antibiotics, Antineoplastic/administration & dosage , Doxorubicin/administration & dosage , Luminescent Agents/chemistry , Nanoconjugates/chemistry , Neoplasms/diagnosis , Neoplasms/therapy , Polymethacrylic Acids/chemistry , Acrylamides/therapeutic use , Animals , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/therapeutic use , Doxorubicin/therapeutic use , Drug Liberation , Female , Humans , Hydrogen-Ion Concentration , Hyperthermia, Induced/methods , Luminescent Agents/therapeutic use , Mice , Mice, Inbred BALB C , Nanoconjugates/therapeutic use , Nanoconjugates/ultrastructure , Optical Imaging , Photochemotherapy/methods , Polymethacrylic Acids/therapeutic use , Temperature , Theranostic Nanomedicine/methods , Tomography, X-Ray Computed
14.
PLoS One ; 9(2): e87585, 2014.
Article in English | MEDLINE | ID: mdl-24503695

ABSTRACT

BACKGROUND: The purpose of this study was to evaluate the activity of Selective Inhibitors of Nuclear Export (SINE) compounds that inhibit the function of the nuclear export protein Exportin 1 (XPO1/CRM1) against canine tumor cell lines and perform a Phase I clinical trial of KPT-335 in dogs with spontaneous cancer to provide a preliminary assessment of biologic activity and tolerability. METHODS AND FINDINGS: Canine tumor cell lines derived from non-Hodgkin lymphoma (NHL), mast cell tumor, melanoma and osteosarcoma exhibited growth inhibition and apoptosis in response to nanomolar concentrations of SINE compounds; NHL cells were particularly sensitive with IC50 concentrations ranging from 2-42 nM. A Phase I clinical trial of KPT-335 was performed in 17 dogs with NHL (naive or relapsed), mast cell tumor or osteosarcoma. The maximum tolerated dose was 1.75 mg/kg given orally twice/week (Monday/Thursday) although biologic activity was observed at 1 mg/kg. Clinical benefit (CB) including partial response to therapy (PR, n = 2) and stable disease (SD, n = 7) was observed in 9/14 dogs with NHL with a median time to progression (TTP) for responders of 66 days (range 35-256 days). A dose expansion study was performed in 6 dogs with NHL given 1.5 mg/kg KPT-335 Monday/Wednesday/Friday; CB was observed in 4/6 dogs with a median TTP for responders of 83 days (range 35-354 days). Toxicities were primarily gastrointestinal consisting of anorexia, weight loss, vomiting and diarrhea and were manageable with supportive care, dose modulation and administration of low dose prednisone; hepatotoxicity, anorexia and weight loss were the dose limiting toxicities. CONCLUSIONS: This study provides evidence that the novel orally bioavailable XPO1 inhibitor KPT-335 is safe and exhibits activity in a relevant, spontaneous large animal model of cancer. Data from this study provides critical new information that lays the groundwork for evaluation of SINE compounds in human cancer.


Subject(s)
Acrylamides/pharmacokinetics , Acrylamides/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Nucleus/metabolism , Drug Evaluation, Preclinical , Hydrazines/pharmacokinetics , Hydrazines/therapeutic use , Neoplasms/drug therapy , Neoplasms/veterinary , Acrylamides/administration & dosage , Acrylamides/adverse effects , Active Transport, Cell Nucleus/drug effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biological Availability , Cell Line, Tumor , Cell Nucleus/drug effects , Dogs , Dose-Response Relationship, Drug , Female , Humans , Hydrazines/administration & dosage , Hydrazines/adverse effects , Inhibitory Concentration 50 , Male , Quality of Life
15.
Nucl Med Biol ; 41(3): 282-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24461626

ABSTRACT

INTRODUCTION: The treatment of prostate cancer using a radiotherapeutic (90)Y labeled N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer can be enhanced with localized tumor hyperthermia. An (111)In labeled HPMA copolymer system for single photon emission computerized tomography (SPECT) was developed to observe the biodistribution changes associated with hyperthermia. Efficacy studies were conducted in prostate tumor bearing mice using the (90)Y HPMA copolymer with hyperthermia. METHODS: HPMA copolymers containing 1, 4, 7, 10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) were synthesized by reversible addition-fragmentation transfer (RAFT) copolymerization and subsequently labeled with either (111)In for imaging or (90)Y for efficacy studies. Radiolabel stability was characterized in vitro with mouse serum. Imaging and efficacy studies were conducted in DU145 prostate tumor bearing mice. Imaging was performed using single photon emission computerized tomography (SPECT). Localized mild tumor hyperthermia was achieved by plasmonic photothermal therapy using gold nanorods. RESULTS: HPMA copolymer-DOTA conjugates demonstrated efficient labeling and stability for both radionuclides. Imaging analysis showed a marked increase of radiolabeled copolymer within the hyperthermia treated prostate tumors, with no significant accumulation in non-targeted tissues. The greatest reduction in tumor growth was observed in the hyperthermia treated tumors with (90)Y HPMA copolymer conjugates. Histological analysis confirmed treatment efficacy and safety. CONCLUSION: HPMA copolymer-DOTA conjugates radiolabeled with both the imaging and treatment radioisotopes, when combined with hyperthermia can serve as an image guided approach for efficacious treatment of prostate tumors.


Subject(s)
Acrylamides/chemistry , Acrylamides/therapeutic use , Gold/chemistry , Gold/therapeutic use , Hyperthermia, Induced , Nanotubes , Prostatic Neoplasms/radiotherapy , Acrylamides/pharmacokinetics , Animals , Cell Line, Tumor , Female , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Male , Mice , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Tomography, Emission-Computed, Single-Photon , Yttrium Radioisotopes/therapeutic use
16.
J Alzheimers Dis ; 34(1): 203-13, 2013.
Article in English | MEDLINE | ID: mdl-23186988

ABSTRACT

This study focused on a promising drug candidate, N-[2-(3,4-dimethoxyphenyl)ethyl]-3-phenyl-acrylamide (gx-50), a compound extracted from Sichuan pepper (Zanthoxylum Bungeanum), to determine whether it would be an effective therapeutic for Alzheimer's disease (AD) via biological experiments. In vivo, we determined the pharmacokinetic profile of gx-50 and evaluated the effect of gx-50 on the cognitive abilities of amyloid-ß protein precursor transgenic (AßPP-Tg) mice by Morris water maze testing. In addition, we examined the effects of gx-50 on amyloid-ß (Aß) oligomers in the brains of AßPP-Tg mice by immunohistochemistry. In vitro, we observed a direct effect of gx-50 on Aß oligomers by atomic force microscopy, detected the neuroprotective effects of gx-50 by western blotting and cell apoptosis assays, and measured its effects on intracellular calcium currents by laser confocal microscopy. Experiments in vivo showed that gx-50 could penetrate the blood brain barrier and improve the cognitive abilities of mice. Moreover, gx-50 treatment decreased the accumulation of Aß oligomers in the cerebral cortex. The results in vitro demonstrated that gx-50 could disassemble Aß oligomers, inhibit Aß-induced neuronal apoptosis and apoptotic gene expression, and reduce neuronal calcium toxicity. These results strongly suggest that gx-50 is a potential candidate drug for treating AD.


Subject(s)
Alzheimer Disease/drug therapy , Neuroprotective Agents/therapeutic use , Phytotherapy/methods , Plant Preparations/therapeutic use , Zanthoxylum/chemistry , Acrylamides/pharmacology , Acrylamides/therapeutic use , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis/drug effects , Brain/cytology , Brain/drug effects , Brain/metabolism , Calcium/metabolism , Cells, Cultured , Cognition Disorders/etiology , Cognition Disorders/prevention & control , Disease Models, Animal , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Plant Preparations/pharmacokinetics , Plaque, Amyloid/drug therapy , Presenilin-1/genetics , Rats , Rats, Sprague-Dawley , Time Factors , bcl-2-Associated X Protein/metabolism
17.
Lab Invest ; 92(3): 371-83, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22157722

ABSTRACT

Src has a role in the anoikis resistance in lung adenocarcinomas. We focused on two epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma cell lines, HCC827 (E746-A750 deletion) and H1975 (L858R+T790M), in suspension to elucidate whether suspended lung adenocarcinoma cells are eradicated by long-term treatment with Src tyrosine kinase inhibitors (TKIs). We also examined metastasis-positive lymph nodes from 16 EGFR-mutant lung adenocarcinoma patients for immunohistochemical expression of mutant-specific EGFR. Almost all suspended HCC827 cells underwent apoptosis after 144 h of combination treatment with AZD0530, trichostatin A (TSA), and ABT-263, whereas many suspended H1975 cells survived the treatment. AZD0530 is a Src TKI, TSA is a histone deacetylase inhibitor, and ABT-263 is a Bcl-2 inhibitor. During the therapy, the phosphorylation of EGFR decreased in HCC827 cells and remained stable in H1975 cells. The phosphorylated EGFR of Src TKI-resistant H1975 cells, as well as HCC827 cells, was completely suppressed by the third generation EGFR TKI, WZ4002. Consequently, both the suspended cell lines were almost completely eradicated within 144 h, with the combined therapy of WZ4002, ABT-263, and TSA. Interestingly, treated suspended cells underwent apoptosis to a greater extent than did adherent cells. Intrasinus floating lung adenocarcinoma cells in the lymph nodes expressed a mutant-specific EGFR. These findings suggest that suspended EGFR-mutant lung adenocarcinoma cells depend significantly more on EGFR activation for survival than attached cells do. The tumor cells circulating in vessels, which express mutant-specific EGFR, would be highly susceptible to the combination therapy of WZ4002, ABT-263, and TSA.


Subject(s)
Acrylamides/therapeutic use , Adenocarcinoma/drug therapy , Anoikis/drug effects , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Pyrimidines/therapeutic use , Acrylamides/pharmacology , Adenocarcinoma/genetics , Aged , Aged, 80 and over , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Therapy, Combination , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Lung Neoplasms/genetics , Male , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrimidines/pharmacology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , src-Family Kinases/antagonists & inhibitors
18.
Neuron ; 70(3): 482-94, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21555074

ABSTRACT

Transient receptor potential melastatin-3 (TRPM3) is a broadly expressed Ca(2+)-permeable nonselective cation channel. Previous work has demonstrated robust activation of TRPM3 by the neuroactive steroid pregnenolone sulfate (PS), but its in vivo gating mechanisms and functions remained poorly understood. Here, we provide evidence that TRPM3 functions as a chemo- and thermosensor in the somatosensory system. TRPM3 is molecularly and functionally expressed in a large subset of small-diameter sensory neurons from dorsal root and trigeminal ganglia, and mediates the aversive and nocifensive behavioral responses to PS. Moreover, we demonstrate that TRPM3 is steeply activated by heating and underlies heat sensitivity in a subset of sensory neurons. TRPM3-deficient mice exhibited clear deficits in their avoidance responses to noxious heat and in the development of inflammatory heat hyperalgesia. These experiments reveal an unanticipated role for TRPM3 as a thermosensitive nociceptor channel implicated in the detection of noxious heat.


Subject(s)
Hot Temperature/adverse effects , Hyperalgesia/metabolism , Pain Threshold/physiology , Sensory Receptor Cells/metabolism , TRPM Cation Channels/metabolism , Acrylamides/therapeutic use , Animals , Behavior, Animal/drug effects , Blood Glucose/drug effects , Blood Glucose/genetics , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Capsaicin/pharmacology , Cell Line, Transformed , Disease Models, Animal , Dose-Response Relationship, Drug , Freund's Adjuvant/adverse effects , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , Gene Expression Regulation/radiation effects , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/genetics , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mustard Plant , Nifedipine/pharmacology , Pain Measurement/drug effects , Pain Measurement/methods , Pain Threshold/drug effects , Patch-Clamp Techniques , Plant Oils/pharmacology , Pregnenolone/adverse effects , Sensory Receptor Cells/drug effects , TRPA1 Cation Channel , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , Telemetry/methods , Time Factors , Transfection/methods , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics , Trigeminal Ganglion/cytology
19.
Clin Exp Nephrol ; 15(1): 14-24, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20820840

ABSTRACT

BACKGROUND: Phosphodiesterase type IV (PDEIV) plays an important role in the immune response and inflammation. However, it is well known that classical PDEIV inhibitors have systemic side effects, so the clinical and chronic use of these agents as therapy for glomerulonephritis is difficult. This study was performed to elucidate the anti-nephritic effects of TJN-598, a new chemical compound derived from herbal components, on experimental mesangial proliferative glomerulonephritis. METHODS: We first examined the effects of TJN-598 and captopril on mesangial expansion induced by anti-Thy1 serum in rats. Second, to investigate the effects of TJN-598 and rolipram, which are typical PDEIV inhibitors, on the production of tumor necrosis factor (TNF)-α and transforming growth factor (TGF)-ß1, glomeruli were isolated from rats with anti-Thy1 nephritis and incubated with the test drugs in vitro for 48 h. RESULTS: Treatment with TJN-598 prevented an increase in the mesangial area/total glomerular area, in the number of cells in the glomerular cross section and matrix index. TJN-598 also inhibited the increases in the expression of α-smooth muscle actin, the TGF-ß1-positive area, in the number of ED-1 positive cells and proliferating cell nuclear antigen-positive cells in the glomeruli. Furthermore, administration of TJN-598 inhibited increases in the levels of TGF-ß1 protein derived from glomeruli with anti-Thy-1 nephritis. The addition of both TJN-598 and rolipram to the culture supernatant inhibited both increased expression of TGF-ß1 and increases in levels of TNF-α in glomeruli isolated from rats with anti-Thy1 nephritis in a dose-dependent manner. CONCLUSION: These results suggest that TJN-598, a PDEIV inhibitor, is effective against expansion of mesangial cells, via the suppression of secretion of TGF-ß1 and TNF-α from inflamed glomeruli.


Subject(s)
Acrylamides/therapeutic use , Cyclic Nucleotide Phosphodiesterases, Type 4 , Glomerulonephritis/drug therapy , Glomerulonephritis/immunology , Isoantibodies/immunology , Phosphodiesterase Inhibitors/therapeutic use , Pyridines/therapeutic use , Acrylamides/chemistry , Animals , Disease Models, Animal , Humans , Male , Molecular Structure , Phosphodiesterase Inhibitors/chemistry , Plant Preparations/therapeutic use , Pyridines/chemistry , Rats , Rats, Wistar , Thy-1 Antigens/immunology , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL