Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Clin Transl Sci ; 16(11): 2369-2381, 2023 11.
Article in English | MEDLINE | ID: mdl-37700528

ABSTRACT

Cisplatin treatment is effective against several types of carcinomas. However, it frequently leads to kidney injury, which warrants effective prevention methods. Sodium valproic acid is a prophylactic drug candidate with a high potential for clinical application against cisplatin-induced kidney injury. Therefore, in this study, we aimed to elucidate the mechanism underlying the prophylactic effect of valproic acid on cisplatin-induced kidney injury in a mouse model and HK2 and PODO cells with cisplatin-induced toxicity. In the mouse model of cisplatin-induced kidney injury, various renal function parameters and tubular damage scores were worsened by cisplatin, but they were significantly improved upon combination with valproic acid. No difference was observed in cisplatin accumulation between the cisplatin-treated and valproic acid-treated groups in whole blood and the kidneys. The mRNA expression levels of proximal tubular damage markers, apoptosis markers, and inflammatory cytokines significantly increased in the cisplatin group 72 h after cisplatin administration but significantly decreased upon combination with valproic acid. In HK2 cells, a human proximal tubular cell line, the cisplatin-induced decrease in cell viability was significantly suppressed by co-treatment with valproic acid. Valproic acid may inhibit cisplatin-induced kidney injury by suppressing apoptosis, inflammatory responses, and glomerular damage throughout the kidneys by suppressing proximal tubular cell damage. However, prospective controlled trials need to evaluate these findings before their practical application.


Subject(s)
Acute Kidney Injury , Cisplatin , Mice , Animals , Humans , Cisplatin/toxicity , Valproic Acid/pharmacology , Prospective Studies , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Kidney , Apoptosis , Mice, Inbred C57BL
2.
J Clin Pharmacol ; 63(4): 473-479, 2023 04.
Article in English | MEDLINE | ID: mdl-36453166

ABSTRACT

Myasthenia gravis (MG) is a rare but fatal adverse event of immune checkpoint inhibitors (ICIs). We assessed whether patient characteristics differed between those with ICI-related myasthenia gravis and those with idiopathic myasthenia gravis. Reports from the US Food and Drug Administration Adverse Event Reporting System were analyzed. Multivariate analyses were conducted to evaluate the associations between age, sex, and ICI treatment and the reporting rate of myasthenia gravis. Among 5 464 099 cases between 2011 and 2019, 53 447 were treated with ICIs. Myasthenia gravis was reported more often in ICI users. Multiple logistic regression analyses showed that the reporting rate of ICI-related myasthenia gravis did not differ significantly between men and women; however, it was higher in older people than in younger people (adjusted odds ratio, 2.4 [95%CI, 1.84-3.13]). We also investigated useful signs for the early detection of myositis and myocarditis, which are fatal when overlapping with ICI-related myasthenia gravis. Patients with elevated serum creatine kinase or troponin levels were more likely to have concurrent myositis and myocarditis. Unlike idiopathic myasthenia gravis, there was no sex difference in the development of ICI-related myasthenia gravis, which may be more common in older people. Considering the physiological muscle weakness that occurs in the elderly, it may be necessary to monitor ICI-related myasthenia gravis more closely in older people.


Subject(s)
Myasthenia Gravis , Myocarditis , Myositis , Male , United States , Humans , Female , Aged , Immune Checkpoint Inhibitors/adverse effects , United States Food and Drug Administration , Myocarditis/chemically induced , Myocarditis/drug therapy , Myasthenia Gravis/chemically induced , Myasthenia Gravis/drug therapy , Myositis/chemically induced , Myositis/drug therapy
3.
Clin Transl Sci ; 15(7): 1664-1675, 2022 07.
Article in English | MEDLINE | ID: mdl-35445533

ABSTRACT

Cisplatin is effective against many types of carcinoma. However, a high rate of renal damage is a clinical problem. Thus, there is a need to establish a method to prevent it. Although various compounds have been reported to be effective against cisplatin-induced renal injury, there are no examples of their clinical application. Therefore, we attempted to search for prophylactic agents with a high potential for clinical application. We used Cascade Eye to identify genes that are altered during cisplatin-induced renal injury, Library of Integrated Network-based Cellular Signatures (LINCS) to identify drugs that inhibit changes in gene expression, and a large database of spontaneous adverse drug reaction reports to identify drugs that could prevent cisplatin-induced kidney injury in clinical practice. In total, 10 candidate drugs were identified. Using the US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS), we identified drugs that reduce cisplatin-induced kidney injury. Fenofibrate was selected as a candidate drug to prevent cisplatin-induced kidney injury based on the FAERS analysis. A model was used to evaluate the efficacy of fenofibrate against cisplatin-induced renal injury. Studies using HK2 cells and mouse models showed that fenofibrate significantly inhibited cisplatin-induced renal injury but did not inhibit the antitumor effect of cisplatin. Fenofibrate is a candidate prophylactic drug with high clinical applicability for cisplatin-induced renal injury. Analysis of data from multiple big databases will improve the search for novel prophylactic drugs with high clinical applicability. For the practical application of these findings, evaluation in prospective controlled trials is necessary.


Subject(s)
Acute Kidney Injury , Fenofibrate , Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Animals , Cisplatin/adverse effects , Data Analysis , Fenofibrate/pharmacology , Kidney , Mice , Prospective Studies
4.
Oncologist ; 27(7): e554-e560, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35325241

ABSTRACT

BACKGROUND: Rash eruptions are a common side-effect of pemetrexed, for which the administration of 8 mg/day of dexamethasone for 3 days from the day preceding pemetrexed administration is recommended. This study aimed to prospectively assess the effectiveness of prophylactic administration of low-dose dexamethasone for pemetrexed-induced rashes. METHODS: This single-arm, phase II study recruited patients with non-squamous non-small cell lung cancer and malignant pleural mesothelioma scheduled to receive chemotherapy including pemetrexed. Patients received 2 mg of dexamethasone daily from days 2 to 6 after chemotherapy with pemetrexed. The primary endpoint was the 3-week incidence of rash eruptions. RESULTS: Twenty-five patients were enrolled between September 2017 and May 2019. The incidence of rash after 3 weeks was 16.7%. Rashes erupted mainly on the upper half of the body, such as the chest and neck, and were of grades 1 and 2 in 2 patients each. No rashes of grade 3 or higher were observed, and there were no adverse events associated with additional corticosteroids. CONCLUSION: Prophylactic administration of low-dose dexamethasone for 5 days from the day after pemetrexed administration resulted in a milder incidence and severity of rash. These findings may provide a standard preventative strategy for pemetrexed-induced rashes. (Trial identifier: UMIN000025666).


Subject(s)
Carcinoma, Non-Small-Cell Lung , Dexamethasone , Exanthema , Lung Neoplasms , Mesothelioma, Malignant , Pemetrexed , Adrenal Cortex Hormones/therapeutic use , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin , Dexamethasone/therapeutic use , Exanthema/chemically induced , Exanthema/drug therapy , Exanthema/prevention & control , Humans , Lung Neoplasms/complications , Lung Neoplasms/drug therapy , Mesothelioma, Malignant/complications , Mesothelioma, Malignant/drug therapy , Pemetrexed/adverse effects
5.
Cancer Med ; 10(1): 164-172, 2021 01.
Article in English | MEDLINE | ID: mdl-33231381

ABSTRACT

BACKGROUND: In patients treated with bevacizumab, hypertension may be a biomarker of therapeutic efficacy. However, it is not clear whether drugs that control blood pressure influence bevacizumab's efficacy. In this study, we investigated drugs that may affect hypertension in bevacizumab-treated patients and examined the impact on the therapeutic effect. PATIENTS AND METHODS: We analyzed 3,724,555 reports from the third quarter of 2010 to the second quarter of 2015. All data were obtained from the Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) analysis. In this retrospective cohort study, we investigated a total of 58 patients diagnosed with colorectal cancer and treated for the first time with bevacizumab containing XELOX or mFOLFOX6 at The University of Tokushima Hospital between January 2010 and December 2015. The effect of the treatment was evaluated according to Response Evaluation Criteria in Solid Tumors version 1.0. Thereafter, the effect was confirmed using Gene Expression Omnibus (GEO) and cultured cells. RESULTS: There are few reports in FAERS of hypertension in patients treated with omeprazole on bevacizumab. Based on the chart review, patients who used proton pump inhibitors (PPI) had a lower response to treatment than those who did not (response rate: 25% vs 50%). Furthermore, experiments on GEO and cell lines suggested that induction of vascular endothelial growth factor (VEGF) gene expression by PPIs is the cause of the reduced therapeutic effect. CONCLUSION: PPIs prevent hypertension in bevacizumab-treated patients but may reduce bevacizumab's anti-tumoral effects by inducing VEGF expression.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Antihypertensive Agents/therapeutic use , Antineoplastic Agents, Immunological/adverse effects , Bevacizumab/adverse effects , Blood Pressure/drug effects , Colorectal Neoplasms/drug therapy , Hypertension/prevention & control , Proton Pump Inhibitors/therapeutic use , Adult , Adverse Drug Reaction Reporting Systems , Aged , Aged, 80 and over , Antihypertensive Agents/adverse effects , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Databases, Factual , Drug Interactions , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypertension/chemically induced , Hypertension/diagnosis , Hypertension/physiopathology , Japan , Male , Middle Aged , Proton Pump Inhibitors/adverse effects , Retrospective Studies , Treatment Outcome , United States , United States Food and Drug Administration , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
6.
Sci Rep ; 10(1): 13773, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792640

ABSTRACT

Immune checkpoint inhibitors (ICIs) elicit antitumour effects by activating the host immunity and cause immune-related adverse events (irAEs). ICI-related interstitial lung disease (ICI-ILD) is a fatal irAE that is difficult to treat; moreover, its incidence is relatively higher in patients with lung cancer. Therefore, early ICI-ILD detection and intervention are important for patient safety. However, a risk assessment method for ICI-ILD has not been established and the prediction of ICI-ILD occurrence is difficult. The aim of our study was to identify the risk factors associated with ICI-ILD. To this end, we retrospectively analysed 102 patients with lung cancer who first received ICI and completed the treatment between April 2016 and December 2019 at Tokushima University Hospital. Nineteen patients had all grades of ICI-ILD and 10 had grade ≥ 3 ICI-ILD. The 30-day mortality rate of patients with grade ≥ 3 ICI-ILD was the highest among all patients (P < 0.01). The multivariate logistic analysis indicated that the performance status ≥ 2 alone and both performance status ≥ 2 and ≥ 50 pack-year were independent risk factors of ICI-ILD of grade ≥ 3 and all grades, respectively. Overall, our study provides insights to predict ICI-ILD occurrence.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/adverse effects , Lung Diseases, Interstitial/chemically induced , Lung Diseases, Interstitial/diagnosis , Lung Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Early Diagnosis , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Lung Diseases, Interstitial/pathology , Male , Middle Aged , Nivolumab/adverse effects , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Retrospective Studies , Risk Assessment , Risk Factors
7.
Biol Pharm Bull ; 42(11): 1839-1845, 2019.
Article in English | MEDLINE | ID: mdl-31685767

ABSTRACT

Uridine 5'-diphospho-glucuronosyltransferase (UGT), a metabolic enzyme of irinotecan active metabolite, has two genetic polymorphisms (UGT1A1*6 and UGT1A1*28). In UGT1A1 homozygous or heterozygous patients, metabolism is delayed and the risk of developing adverse effects is increased, and therefore, dose reduction of irinotecan is considered. However, the specific dose reduction rate of irinotecan for heterozygous patients is uncertain. We studied the necessity of irinotecan dose reduction and its optimal dose in UGT1A1 heterozygous patients with lung cancer. Patients with lung cancer treated with irinotecan in the Tokushima University Hospital or Tokushima Municipal Hospital were included in this study. The dose of irinotecan was evaluated based on the relative dose intensity (RDI). The time to treatment failure (TTF) was defined as the period until treatment change, death, or progressive disease based on response evaluation criteria of solid tumors. We targeted 31 patients treated with irinotecan: 12 wild types (WT), 14 heterozygotes, and 1 complex heterozygote and 4 homozygotes. There was no significant difference in the TTF, but the mean RDI during the entire treatment period was significantly different in the wild type (79%), heterozygous (62%), and complex heterozygous and homozygous groups (46%). In addition, the proportion of patients who completed treatment without dose reduction in the WT group tended to be higher than that in the other groups. For lung cancer patients with UGT1A1 heterozygote types who start irinotecan therapy, reducing the initial dose by approximately 20% might be a safer chemotherapy without decreasing the therapeutic effect.


Subject(s)
Glucuronosyltransferase/genetics , Irinotecan/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Aged , Dose-Response Relationship, Drug , Female , Genotype , Glucuronosyltransferase/metabolism , Heterozygote , Homozygote , Humans , Irinotecan/adverse effects , Lung Neoplasms/genetics , Male , Middle Aged , Polymorphism, Genetic
8.
Cancer Chemother Pharmacol ; 84(5): 1107-1114, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31502114

ABSTRACT

PURPOSE: Epirubicin and cyclophosphamide (EC) therapy, a major chemotherapy for patients with early-stage breast cancer, has a low risk (< 10%) of febrile neutropenia (FN). However, data used in reports on the incidence rate of FN were derived primarily from non-Asian populations. In this study, we investigated the FN incidence rate using EC therapy among Japanese patients with breast cancer and evaluated the significance of prophylactic administration of granulocyte colony-stimulating factor (G-CSF). METHODS: We evaluated medical records of patients with early-stage breast cancer who had been treated with EC therapy as neoadjuvant or adjuvant therapy between November 2014 and July 2018. RESULTS: The incidence rate of FN was 23.9%. In patients who received G-CSF as primary prophylaxis, FN expression was completely suppressed. The incidence rate of severe leucopenia/neutropenia, emergency hospitalization, and the use of antimicrobial agents were low in patients receiving primary prophylaxis with G-CSF compared with those not receiving G-CSF (27.3% vs. 64.8%, 9.1% vs. 27.3%, and 27.3% vs. 71.6%, respectively). Furthermore, in all patients who received primary prophylaxis with G-CSF, a relative dose intensity > 85% using EC therapy was maintained. CONCLUSION: The incidence of FN in EC therapy among Japanese patients was higher than expected, EC therapy appears to be a high-risk chemotherapy for FN, and prophylactic administration of G-CSF is recommended. Maintaining high therapeutic intensity is associated with a positive prognosis for patients with early breast cancer, and prophylactic administration of G-CSF is likely to be beneficial in treatment involving EC therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Febrile Neutropenia/prevention & control , Granulocyte Colony-Stimulating Factor/administration & dosage , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/pathology , Cyclophosphamide/administration & dosage , Epirubicin/administration & dosage , Febrile Neutropenia/chemically induced , Febrile Neutropenia/epidemiology , Female , Humans , Incidence , Japan , Middle Aged , Neoplasm Staging , Retrospective Studies
9.
Yakugaku Zasshi ; 138(11): 1363-1370, 2018.
Article in Japanese | MEDLINE | ID: mdl-30381644

ABSTRACT

Cancer chemotherapy has a high frequency of side effects, and patients often experience adverse health effects. This review focuses on risk factors and supportive care for the prevention of chemotherapy side effects. 1) Drug-induced interstitial lung disease (DILD) is one of the serious adverse events associated with chemotherapy, and this retrospective study investigated the risk of DILD in Japanese patients with lung cancer. Among the 459 patients who received lung cancer chemotherapy from April 2007 to March 2013, 33 (7.2%) developed chemotherapy-associated DILD. Preexisting interstitial lung disease was a risk factor for DILD [odds ratio (OR)=5.38; 95% confidence interval (CI)=2.47-11.73]. Early death was observed in 10 of the 33 patients who developed DILD. Epidermal growth factor receptor-tyrosine kinase inhibitor use (OR=9.26; 95%CI=1.05-81.96) and two or more prior chemotherapy regimens (OR=6.95; 95%CI=1.14-42.35) were identified as poor prognostic factors. 2) The incidence of pemetrexed-induced rash is high. The aim of this retrospective study was to evaluate the efficacy of corticosteroids for pemetrexed-induced rash. Rash developed in 26.9% of patients who received pemetrexed between April 2009 and March 2014. Supplementation with dexamethasone (≥1.5 mg) on days 2 and 3 significantly reduced the incidence of rash compared with no supplementary corticosteroids (39.4% vs. 17.8%, p<0.05). Increasing the corticosteroid dose had no additional effect on pemetrexed-induced rash development. These results suggest that supplementary corticosteroids may prevent pemetrexed-induced rash, and low-dose corticosteroids are sufficient for such prevention.


Subject(s)
Antineoplastic Agents/adverse effects , Drug-Related Side Effects and Adverse Reactions/prevention & control , Lung Diseases, Interstitial/chemically induced , Lung Diseases, Interstitial/prevention & control , Lung Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Dexamethasone/administration & dosage , Exanthema/chemically induced , Exanthema/drug therapy , Female , Glucocorticoids/administration & dosage , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors
10.
Pharmacology ; 102(5-6): 287-299, 2018.
Article in English | MEDLINE | ID: mdl-30253416

ABSTRACT

BACKGROUND/AIMS: We have reported that nitrosonifedipine (NO-NIF), a photodegradation product of nifedipine, has strong antioxidant and endothelial protective effects, and can suppress several cardiovascular diseases in animal models. The objective of the present study was to investigate the effects of NO-NIF on aortic aneurysm formation. METHODS: The mice were infused with ß-aminopropionitrile for 2 weeks and angiotensin II for 6 weeks to induce aortic aneurysm formation. The oxidative stress was measured by dihydroethidium staining and nitrotyrosine staining. The expressions of inflammation-related genes were assessed by quantitative real-time PCR and immunohistochemical staining. To clarify the mechanisms of how NO-NIF suppresses vascular cell adhesion molecule (VCAM)-1, endothelial cells were used in in vitro system. RESULTS: NO-NIF suppressed pharmacologically induced the aortic aneurysm formation and aortic expansion without blood pressure changes. NO-NIF suppressed elastin degradation and matrix metalloproteinase-2 mRNA expression. NO-NIF suppressed the reactive oxygen species-cyclophilin A positive feedback loop. Upregulated mRNA expressions of inflammation-related genes and endothelial VCAM-1 were suppressed by NO-NIF co-treatment in aortae. CONCLUSION: NO-NIF has the potential to be a new, nifedipine-derived therapeutic drug for suppressing aortic aneurysm formation by directly improving aortic structure with its strong ability to reduce oxidative stress and inflammation.


Subject(s)
Aortic Aneurysm/drug therapy , Nifedipine/analogs & derivatives , Nitroso Compounds/pharmacology , Aminopropionitrile/administration & dosage , Angiotensin II/administration & dosage , Animals , Antigens, Differentiation/metabolism , Antioxidants/pharmacology , Aortic Aneurysm/chemically induced , Aortic Aneurysm/metabolism , Chemokine CCL2/metabolism , Cyclophilins/metabolism , Disease Models, Animal , Elastin/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Matrix Metalloproteinase 2/metabolism , Mice , Nifedipine/pharmacology , Oxidative Stress/drug effects , Photolysis , Reactive Oxygen Species/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
11.
Article in English | MEDLINE | ID: mdl-28748102

ABSTRACT

BACKGROUND: In chemotherapy, the full round of treatment must be completed as scheduled to achieve the strongest therapeutic effect. However, peripheral neuropathy, a severe side effect of the chemotherapeutic agent paclitaxel, can force the premature discontinuation of treatment. As some kampo practitioners have suggested that it may be possible to counteract such side effects, we analyzed the effects of Kamishoyosan, Shakuyakukanzoto, and Goshajinkigan in an in vitro model of paclitaxel-induced peripheral neuropathy. METHODS: Paclitaxel-treated PC12 cells were assessed for neurite length and performed Western blot analysis for growth-associated protein-43 (GAP-43) and light neurofilament protein (NF-L) levels in the presence of nerve growth factor (NGF); they were re-assessed, with additional testing for acetylcholinesterase levels, after application of one of the kampo. We also compared phosphorylation of extracellular signal-regulated kinase (Erk)1/2 and Akt via Western blot analysis. About effect of kampo to anticancer efficacy, we confirmed cell cytotoxicity in A549 cells using MTT assay. RESULTS: Addition of Kamishoyosan or Shakuyakukanzoto, but not Goshajinkigan, significantly improved neurite length and GAP-43 and NF-L levels from paclitaxel-treated PC12 cells, relative to those of only NGF-treated PC12 cells. The promoting effect of Kamishoyosan and Shakuyakukanzoto in neurite outgrowth is confirmed when NGF promoted neurite outgrowth, and it was inhibited partially when Erk1/2 and Akt were blocked by Erk1/2 inhibitor or Akt inhibitor alone. Furthermore, neurite outgrowth induced by TJ24 and TJ68 was inhibited more strongly when Erk1/2 inhibitor and Akt inhibitor were treated at the same time. NGF with Kamishoyosan or Shakuyakukanzoto promoted the proportion of phosphorylated Erk1/2 and phosphorylated Akt compare with NGF only. On the other hand, Kamishoyosan or Shakuyakukanzoto didn't influence cytotoxicity of paclitaxel in A549 cells. CONCLUSIONS: Kamishoyosan or Shakuyakukanzoto promotes neurite outgrowth with NGF via increasing the proportion of phosphorylated Erk1/2 and phosphorylated Akt in PC12 cells. The effect applies to recovery from paclitaxel-induced axonal involvement and might promote recovery from paclitaxel-induced neuropathy without influence of anticancer effect of paclitaxel.

12.
Biol Pharm Bull ; 38(11): 1752-6, 2015.
Article in English | MEDLINE | ID: mdl-26521826

ABSTRACT

Pemetrexed, a chemotherapeutic drug, is highly active in non-small cell lung cancer and malignant pleural mesothelioma. Unfortunately, rashes are more commonly associated with pemetrexed than other chemotherapies, and it is recommended that patients receive corticosteroids (8 mg/d of dexamethasone) for 3 d, including the day of pemetrexed administration (day 1). However, the efficacy of corticosteroids in this context has not been fully verified. In this retrospective study, we evaluated the medical records of 78 patients who received pemetrexed between April 2009 and March 2014, to confirm whether supplementary corticosteroids prevented rash development. The incidence of rash was lower in the 47 patients who received supplementary corticosteroids (after day 1) compared with the incidence among the 31 patients who did not receive supplementary corticosteroids (19.1% vs. 38.7%). The average cutoff dosage of supplementary corticosteroids on day 2 and day 3 was 1.5 mg/d of dexamethasone, as calculated using the receiver operating characteristic curve, and the odds ratio was 0.33 (95% confidence interval: 0.12-0.94). Administration of ≥1.5 mg of corticosteroids on day 2 and day 3 significantly reduced the severity of the rash compared to no supplementary treatment (grades 2/3, 13.3% vs. 33.3%, p<0.05). However, increasing the dose of corticosteroids had no additional effect on rash development. These results suggest that ≥1.5 mg of supplementary dexamethasone on day 2 and day 3 (in addition to day 1) may be necessary for preventing pemetrexed-induced rash, but high doses of dexamethasone (e.g., 8 mg/d) are unnecessary.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Dexamethasone/therapeutic use , Exanthema/prevention & control , Neoplasms/drug therapy , Pemetrexed/adverse effects , Adrenal Cortex Hormones/administration & dosage , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Area Under Curve , Dexamethasone/administration & dosage , Exanthema/chemically induced , Female , Humans , Male , Middle Aged , Odds Ratio , Pemetrexed/therapeutic use , ROC Curve , Retrospective Studies , Severity of Illness Index , Treatment Outcome
13.
Ann Pharmacother ; 49(4): 398-404, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25565405

ABSTRACT

BACKGROUND: Drug-induced interstitial lung disease (DILD) is generally a serious adverse effect and almost always necessitates the discontinuation of the offending drug. Cancer pharmacotherapy is strongly associated with DILD, and the risk of DILD has been suggested to be higher in patients with lung cancer because of preexisting pneumonic disease. OBJECTIVE: The aim of this retrospective study was to identify the risk factors and prognostic factors for early death from interstitial lung disease (ILD) induced by chemotherapy for lung cancer. METHODS: The medical records of 459 patients who underwent chemotherapy for lung cancer between April 2007 and March 2013 were analyzed with regard to patient background and DILD development, initial symptoms, and prognosis. RESULTS: A total of 33 patients (7.2%) developed chemotherapy-induced ILD. The most frequently observed initial symptom was dyspnea (94.3%). Preexisting ILD was identified as a risk factor for DILD (odds ratio [OR] = 5.38; 95% CI = 2.47-11.73; P < 0.01). Among the 33 patients who developed DILD, 10 patients suffered an early death despite steroid therapy. Poor prognostic factors included epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) use (OR = 9.26; 95% CI = 1.05-82.0; P < 0.05) and 2 or more prior chemotherapy regimens (OR = 6.95; 95% CI = 1.14-42.3; P < 0.05). CONCLUSIONS: Many lung cancer patients have coexisting ILD, and these patients have a high risk of developing chemotherapy-induced ILD. In addition, patients with DILD who underwent EGFR-TKI therapy and 2 or more prior chemotherapy regimens had a higher risk of fatal outcome.


Subject(s)
Antineoplastic Agents/adverse effects , Lung Diseases, Interstitial/chemically induced , Lung Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Female , Humans , Lung Diseases, Interstitial/etiology , Male , Middle Aged , Odds Ratio , Prognosis , Retrospective Studies , Risk Factors
14.
Yakugaku Zasshi ; 134(6): 715-9, 2014.
Article in Japanese | MEDLINE | ID: mdl-24882646

ABSTRACT

Oxidative stress is a key factor involved in the pathogenesis and progression of cardiovascular disease (CVD) and chronic kidney disease (CKD). Reactive oxygen species (ROS), produced as a result of redox reactions in various cells, have been recognized as key chemical mediators causing cellular damage and organ dysfunction in CVD and CKD. Nifedipine, a well-known calcium channel blocker, is extremely sensitive to light which gets converted to its nitroso analog, nitrosonifedipine (NO-NIF) in the presence of ultraviolet and visible light. The so formed NO-NIF blocks calcium channel quite weakly compared to that of nifedipine. However, we elucidated for the first time that NO-NIF is converted to NO-NIF radical which acquires extremely strong antioxidant property via reaction with unsaturated fatty acid or endothelial cells. We have already reported that NO-NIF reduces the cytotoxicity of cumene hydroperoxide, which hampers the integrity of cell membrane through oxidative stress, in endothelial cells. Additionally, we demonstrated that NO-NIF restored acetylcholine-responsive vascular relaxation and suppressed intercellular adhesion molecule-1 expression in the aorta of N(ω)-nitro-L-arginine methyl ester-treated rats, a model of vascular endothelial dysfunction. Recently, we reported that NO-NIF ameliorates angiotensin II-induced vascular remodeling via antioxidative effects in vivo and in vitro. These observations point towards the plausible, unique role of NO-NIF as a novel antioxidant which improves vascular dysfunction for overcoming CVD and CKD and the same has been highlighted in this review.


Subject(s)
Cardiovascular Diseases/drug therapy , Oxidative Stress , Renal Insufficiency, Chronic/drug therapy , Animals , Cardiovascular Diseases/metabolism , Humans , Nifedipine/analogs & derivatives , Nifedipine/metabolism , Nitric Oxide/metabolism , Nitroso Compounds/metabolism , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/metabolism
15.
J Clin Med Res ; 6(4): 252-60, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24883150

ABSTRACT

BACKGROUND: Peripheral neuropathy is a well-known side effect of vincristine (VCR), a microtubule inhibitor used for R-CHOP or R-CHOP-like (namely R-CVP and R-THP-COP) regimens. Previous studies have shown that both the total dose of VCR and the number of treatment cycles are related to the incidence of VCR-induced peripheral neuropathy (VIPN). However, VIPN will also occur during the first treatment cycle regardless of the total dose of VCR or number of treatment cycles (early-onset VIPN). There is little information about early-onset VIPN, and it is difficult to predict. The present study's goal was to identify risk factors for early-onset VIPN. METHODS: We analyzed the case records of patients who had their first administration of an R-CHOP or R-CHOP-like regimen between April 2008 and August 2013 at Tokushima University Hospital in Tokushima, Japan. To identify the risk factors for early-onset VIPN, we performed univariate and multivariate logistic regression analyses. RESULTS: Forty-one patients underwent an R-CHOP or R-CHOP-like regimen for the first time at Tokushima University Hospital between April 2008 and August 2013, and 14 patients had grade 1 or higher early-onset VIPN. A univariate analysis revealed that age, the dose of VCR and the concomitant use of aprepitant appeared to be the risk factors of early-onset VIPN. In our calculation using receiver-operator characteristics curves, the cut-off value for patient age was 65 years and that of the dose of VCR was 1.9 mg. A multivariate analysis revealed that VCR dose ≥ 1.9 mg and the concomitant use of the antiemetic aprepitant were independent risk factors for early-onset VIPN. CONCLUSIONS: Our present study showed that the patients who had VCR dose ≥ 1.9 mg and the concomitant use of aprepitant had the risk for early-onset VIPN. This suggests that it is important to use aprepitant in light of the risk of early-onset VIPN and the benefit of aprepitant's antiemetic effect in R-CHOP and R-CHOP-like regimens.

16.
PLoS One ; 9(1): e86335, 2014.
Article in English | MEDLINE | ID: mdl-24489716

ABSTRACT

Diabetic nephropathy (DN) is the major cause of end-stage renal failure. Oxidative stress is implicated in the pathogenesis of DN. Nitrosonifedipine (NO-NIF) is a weak calcium channel blocker that is converted from nifedipine under light exposure. Recently, we reported that NO-NIF has potential as a novel antioxidant with radical scavenging abilities and has the capacity to treat vascular dysfunction by exerting an endothelial protective effect. In the present study, we extended these findings by evaluating the efficacy of NO-NIF against DN and by clarifying the mechanisms of its antioxidative effect. In a model of type 2 DN (established in KKAy mice), NO-NIF administration reduced albuminuria and proteinuria as well as glomerular expansion without affecting glucose metabolism or systolic blood pressure. NO-NIF also suppressed renal and systemic oxidative stress and decreased the expression of intercellular adhesion molecule (ICAM)-1, a marker of endothelial cell injury, in the glomeruli of the KKAy mice. Similarly, NO-NIF reduced albuminuria, oxidative stress, and ICAM-1 expression in endothelial nitric oxide synthase (eNOS) knockout mice. Moreover, NO-NIF suppressed urinary angiotensinogen (AGT) excretion and intrarenal AGT protein expression in proximal tubular cells in the KKAy mice. On the other hand, hyperglycemia-induced mitochondrial superoxide production was not attenuated by NO-NIF in cultured endothelial cells. These findings suggest that NO-NIF prevents the progression of type 2 DN associated with endothelial dysfunction through selective antioxidative effects.


Subject(s)
Antioxidants/therapeutic use , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Nifedipine/analogs & derivatives , Nitroso Compounds/therapeutic use , Animals , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nifedipine/therapeutic use , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects
17.
Naunyn Schmiedebergs Arch Pharmacol ; 386(1): 29-39, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23149861

ABSTRACT

Nifedipine is unstable under light and decomposes to a stable nitroso analog, nitrosonifedipine (NO-NIF). The ability of NO-NIF to block calcium channels is quite weak compared with that of nifedipine. Recently, we have demonstrated that NO-NIF reacts with unsaturated fatty acid leading to generate NO-NIF radical, which acquires radical scavenging activity. However, the effects of NO-NIF on the pathogenesis related with oxidative stress, such as atherosclerosis and hypertension, are unclear. In this study, we investigated the effects of NO-NIF on angiotensin II (Ang II)-induced vascular remodeling. Ang II-induced thickening and fibrosis of aorta were inhibited by NO-NIF in mice. NO-NIF decreased reactive oxygen species (ROS) in the aorta and urinary 8-hydroxy-20-deoxyguanosine. Ang II-stimulated mRNA expressions of p22(phox), CD68, F4/80, monocyte chemoattractant protein-1, and collagen I in the aorta were inhibited by NO-NIF. Moreover, NO-NIF inhibited Ang II-induced cell migration and proliferation of vascular smooth muscle cells (VSMCs). NO-NIF reduced Ang II-induced ROS to the control level detected by dihydroethidium staining and lucigenin chemiluminescence assay in VSMCs. NO-NIF suppressed phosphorylations of Akt and epidermal growth factor receptor induced by Ang II. However, NO-NIF had no effects on intracellular Ca(2+) increase and protein kinase C-δ phosphorylation induced by Ang II in VSMCs. The electron paramagnetic resonance spectra indicated the continuous generation of NO-NIF radical of reaction with cultured VSMCs. These findings suggest that NO-NIF improves Ang II-induced vascular remodeling via the attenuation of oxidative stress.


Subject(s)
Angiotensin II/pharmacology , Antioxidants/pharmacology , Nifedipine/analogs & derivatives , Nitroso Compounds/pharmacology , Oxidative Stress/drug effects , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Calcium/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Fibrosis , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nifedipine/pharmacology , Rats , Reactive Oxygen Species/metabolism
18.
Pharmacology ; 90(5-6): 324-31, 2012.
Article in English | MEDLINE | ID: mdl-23052181

ABSTRACT

BACKGROUND/AIMS: Tumor necrosis factor-α (TNF-α) is known to involve the progression of renal dysfunction through its cytotoxicity and proinflammatory effects such as the induction of intercellular adhesion molecule (ICAM)-1 expression in vascular endothelial cells (ECs). Olmesartan, one of the angiotensin II type 1 receptor blockers (ARBs), has been reported to show protective effects on injured ECs by some causal factors of renal disorder other than angiotensin II. However, the effects of olmesartan on TNF-α-induced glomerular EC damage have not been investigated. In the present study, we investigated the effects of RNH-6270, an active metabolite of olmesartan, on TNF-α-induced human glomerular EC (HGEC) damage to clarify the renoprotective mechanisms of ARBs. METHODS: Cultured HGECs were stimulated by TNF-α, and then cell viability and cytotoxicity were measured by MTT assay and lactate dehydrogenase release assay, respectively. TNF-α-induced oxidative stress was estimated by dihydroethidium assay and lucigenin chemiluminescence assay. ICAM-1 expression and the phosphorylations of mitogen-activated protein kinases were measured using Western blotting assay. RESULTS: RNH-6270 suppressed cell death and the increase in ICAM-1 expression induced by TNF-α via the inhibition of reactive oxygen species in HGECs. CONCLUSION: Our findings suggested that olmesartan might have protective effects against TNF-α-induced glomerular EC dysfunction.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Apoptosis/drug effects , Endothelial Cells/drug effects , Imidazoles/pharmacology , Tetrazoles/pharmacology , Tumor Necrosis Factor-alpha , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Kidney Glomerulus/cytology , Reactive Oxygen Species/metabolism
19.
Yakugaku Zasshi ; 131(8): 1251-7, 2011.
Article in Japanese | MEDLINE | ID: mdl-21804330

ABSTRACT

Bevacizumab is a monoclonal antibody that targets vascular endothelial growth factor (VEGF) for treatment of metastatic colorectal cancer. Recently, much evidence has suggested that bevacizumab-induced hypertension might be predictive of the effect of bevacizumab. The aim of our study is to retrospectively assess the relationship between the onset of hypertension and the activity of bevacizumab in Japanese metastatic colorectal cancer patients. Between July 2007 and December 2010, 36 patients (median age 66 years; 36-81 years) with metastatic colorectal cancer were assigned to receive bevacizumab in combination with either mFOLFOX6 (5-FU, levofolinate and oxaliplatin) or FOLFIRI (5-FU, levofolinate and irinotecan) at the Tokushima University Hospital. A patient who had increase by >20 mmHg in diastolic blood pressure or had increase to >150/100 mmHg or received antihypertensive treatment was defined as hypertensive. The objective response rate (ORR), disease control rate (DCR) and progression-free survival (PFS) were compared between the hypertensive group (n=10) and non-hypertensive group (n=26). ORR and DCR were 60.0% and 100%, respectively, in the hypertensive group and ORR and DCR were 23.1% and 80.8%, respectively, in the non-hypertensive group. These differences were statistically significant (p<0.05). The median PFS tended to be longer in the hypertensive group (65.0 weeks) than in the non-hypertensive group (40.0 weeks). Our data suggested that bevacizumab-induced hypertension may be predictive of the effect of bevacizumab in Japanese metastatic colorectal cancer patients.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Blood Pressure/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/secondary , Hypertension/chemically induced , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Asian People , Bevacizumab , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/etiology , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Forecasting , Humans , Leucovorin/administration & dosage , Male , Middle Aged , Molecular Targeted Therapy , Organoplatinum Compounds/administration & dosage , Retrospective Studies , Treatment Outcome , Vascular Endothelial Growth Factor A/immunology
20.
Neurosci Res ; 53(3): 323-30, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16157407

ABSTRACT

To understand the neuronal cell type-specific expression of Ca2+/calmodulin-dependent protein kinase II (CaM kinase II), we investigated binding proteins that specifically activated the promoter of the alpha isoform of CaM kinase II (alpha CaM kinase II). Proteins that bind the promoter sequence were found in rat brain nuclear extract by electrophoretic mobility shift assay. Then, we screened for binding proteins in a mouse brain cDNA library using the yeast one-hybrid system. Zic2, a Zic family zinc finger transcription factor, was identified as one of the binding proteins. To investigate the effect of Zic2 on the promoter activity, Zic2 cDNA was expressed with a luciferase reporter gene containing a neuronal cell type-specific promoter of alpha CaM kinase II in neuronal and non-neuronal cells. The promoter activity of alpha CaM kinase II was enhanced 1.3-5-fold in cultured neuronal cells by Zic2. The activation was varied among neuronal cell types. Zic2 also increased the promoter activity in non-neuronal cells, although the relative luciferase activites in non-neuronal cells were lower than those in neuronal cell lines. These results indicated that Zic2 was one of the proteins binding to, and regulating the activity of, the promoter of alpha CaM kinase II.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/genetics , Neurons/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cell Line , Cell Line, Tumor , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic/genetics , Humans , Luciferases/genetics , Mice , Neurons/cytology , Regulatory Elements, Transcriptional/genetics , Transcriptional Activation/genetics , Zinc Fingers/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...