Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Front Oncol ; 13: 1216813, 2023.
Article in English | MEDLINE | ID: mdl-37483483

ABSTRACT

Background: Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most common adverse events that can significantly impair the quality of life of patients. Although limb cooling may be beneficial for preventing CIPN, logistical challenges exist in ensuring consistent efficacy and safety. The purpose of this randomized controlled trial is to validate whether limb cooling with strict temperature control can reduce CIPN in patients with breast cancer receiving weekly paclitaxel as a perioperative treatment. Methods: This study is a multicenter, double-blinded, randomized controlled trial. We plan to enroll patients with breast cancer who are scheduled to receive 12 weekly doses of paclitaxel (60 min 80 mg/m2 intravenous infusion) as perioperative chemotherapy. Patients will be randomly divided into the intervention or control groups and undergo limb cooling therapy maintained at a constant temperature of 13°C and 25°C, respectively. The primary endpoint is the proportion of patients who report Patient Neurotoxicity Questionnaire (PNQ) ≥ D in their limbs by the end of the study treatment or at the time of discontinuation. Discussion: The results of this trial will contribute to the establishment of new evidence for limb cooling therapy in the mitigation of CIPN and present a safe and stable cooling device that may be suitable for use in the clinic. Clinical trial registration: https://jrct.niph.go.jp/en-latest-detail/jRCT2032210115, identifier jRCT2032210115.

2.
Clin Colorectal Cancer ; 22(1): 45-52, 2023 03.
Article in English | MEDLINE | ID: mdl-36376197

ABSTRACT

Utilizing real-world data (RWD) for effective clinical implementation is becoming more and more appealing as the cost of drug development rises, especially for patients with rare diseases and rare molecular subtypes for whom conducting randomized controlled trials is challenging. If a regulatory approval methodology based on RWD as an external control group can be established, drug development for rarer fractions can be accelerated by lowering costs and time, as well as reducing physical and emotional burdens on both patients and healthcare professionals. Since 2017, we have been prospectively collecting the clinical data of standard therapies in patients with rare molecular fractions under the SCRUM-Japan Registry platform, which is a qualified registry utilized as external control data for regulatory submission. Based on the results of the phase II TRIUMPH study (UMIN000027887) and the extracted data from the SCRUM-Japan Registry, the pharmaceutical company submitted an application for pertuzumab and trastuzumab in patients with HER2-positive metastatic colorectal cancer in April 2021. Pertuzumab and trastuzumab were approved as expanded indications on March 28, 2022, as 6 cases out of 14 extracted from the SCRUM-Japan Registry were classified and utilized as "evaluation material" under the review process of the Pharmaceuticals and Medical Devices Agency (PMDA). Through the TRIUMPH study and the SCRUM-Japan Registry, we have paved the way for regulatory approval of RWD in Japan. In future, we must define the steps for constructing regulatory-grade registries and the method/process for utilizing RWD by accumulating case experiences.


Subject(s)
Breast Neoplasms , Colorectal Neoplasms , Humans , Female , Receptor, ErbB-2 , Antibodies, Monoclonal, Humanized/therapeutic use , Trastuzumab/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/etiology , Breast Neoplasms/drug therapy
3.
Ther Innov Regul Sci ; 56(2): 220-229, 2022 03.
Article in English | MEDLINE | ID: mdl-34787814

ABSTRACT

BACKGROUND: The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) is undertaking a major revision of ICH E6 Good Clinical Practice (GCP) decided to involve external stakeholders in ICH-GCP renovation. Activities such as surveys and public conferences have taken place in the United States, European Union, and Japan. For stakeholder engagement in Japan, a designated research group conducted a survey of academic stakeholders. METHODS: A total of 105 academic stakeholders from 18 institutions responded to the survey. The research group developed recommendations reflecting the survey results and the opinions from patients and the public. RESULTS: The survey showed the top four principles needing renovation were (i) informed consent (Chapter 2.9, 12.4% of respondents believed it needed renovation), (ii) systems for quality assurance (Chapter 2.13, 9.5%), (iii) information on an investigational product (Chapter 2.4, 5.7%), and (iv) procedures on clinical trial information (Chapter 2.10, 5.7%). The top three sections identified as needing renovation were: (i) informed consent (Chapter 4.8, 27.6%), (ii) monitoring (Chapter 5.18, 22.9%), and (iii) composition, functions, and operations of the ethics committee (Chapter 3.2, 14.3%). Recommendations included clarification of ICH-GCP's scope, proportionality in various aspects of clinical trials, diversity and liquidity of ethics committee members, modernization of informed consent procedures, variations in monitoring, and regulatory grade when using real-world data. CONCLUSION: The recommendations from Japanese investigators and patients have been submitted to the ICH E6 Expert Working Group, which will strengthen the robustness of the GCP renovation.


Subject(s)
Informed Consent , Research Personnel , European Union , Humans , Japan , Surveys and Questionnaires , United States
4.
Nat Med ; 27(11): 1899-1903, 2021 11.
Article in English | MEDLINE | ID: mdl-34764486

ABSTRACT

The applicability of circulating tumor DNA (ctDNA) genotyping to inform enrollment of patients with cancer in clinical trials has not been established. We conducted a phase 2 trial to evaluate the efficacy of pertuzumab plus trastuzumab for metastatic colorectal cancer (mCRC), with human epidermal growth factor receptor 2 (HER2) amplification prospectively confirmed by tumor tissue or ctDNA analysis ( UMIN000027887 ). HER2 amplification was confirmed in tissue and/or ctDNA in 30 patients with mCRC. The study met the primary endpoint with a confirmed objective response rate of 30% in 27 tissue-positive patients and 28% in 25 ctDNA-positive patients, as compared to an objective response rate of 0% in a matched real-world reference population treated with standard-of-care salvage therapy. Post hoc exploratory analyses revealed that baseline ctDNA genotyping of HER2 copy number and concurrent oncogenic alterations adjusted for tumor fraction stratified patients according to efficacy with similar accuracy to tissue genotyping. Decreased ctDNA fraction 3 weeks after treatment initiation associated with therapeutic response. Pertuzumab plus trastuzumab showed similar efficacy in patients with mCRC with HER2 amplification in tissue or ctDNA, showing that ctDNA genotyping can identify patients who benefit from dual-HER2 blockade as well as monitor treatment response. These findings warrant further use of ctDNA genotyping in clinical trials for HER2-amplified mCRC, which might especially benefit patients in first-line treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Circulating Tumor DNA/blood , Colorectal Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Trastuzumab/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Female , Gene Dosage/genetics , Genotyping Techniques , Humans , Male , Middle Aged , Prospective Studies , Translational Research, Biomedical
5.
Cancer Sci ; 112(7): 2563-2577, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33990993

ABSTRACT

Clinical studies intended for regulatory approval must demonstrate the clinical benefits of the drug in a target population. Clinical development of a drug proceeds by stepwise clinical studies; after safety and pharmacokinetics are evaluated and the recommended dosage and administration are determined, efficacy and safety are evaluated in an exploratory manner, and finally clinical benefits are compared with conventional standard therapies. Guidelines for the clinical evaluation of anti-cancer drugs in Japan were established in 1991 and amended in 2006 after molecular-targeted drugs were introduced. Recent progress in the development of drugs acting on the immune system and cancer genomic medicine targeting rare but important molecular subtypes have altered the strategy for development of anti-cancer drugs. It is often difficult to conduct a confirmatory randomized controlled study using overall survival as the primary endpoint in rare molecular subtypes, and the primary evaluation of the efficacy of some drugs and subsequent approval is based on the tumor response. As conducting clinical studies for rare subtypes solely within Japan is difficult, drug development needs to be conducted within a global study. However, this requires robust monitoring to detect possible ethnic differences in pharmacokinetics and drug efficacy. Development using the conditional approval system for drugs enforced in 2020 may be considered, when clinical utility is evaluated based on surrogate endpoints. Because of these changes, we have revised the guidelines for the clinical evaluation of anti-cancer drugs in Japan. To promote global development of anti-cancer drugs involving Japan, the guidelines have been translated into English.


Subject(s)
Antineoplastic Agents/therapeutic use , Clinical Studies as Topic/standards , Antineoplastic Agents/pharmacology , Drug Development/organization & administration , Drug Development/standards , Humans , Japan , Neoplasms/drug therapy , Rare Diseases/drug therapy , Treatment Outcome
6.
Jpn J Clin Oncol ; 51(6): 879-885, 2021 May 28.
Article in English | MEDLINE | ID: mdl-33561262

ABSTRACT

OBJECTIVE: We evaluated the efficacy and safety of first-line S-1 plus cisplatin in combination with cetuximab for Japanese patients with advanced gastric cancer, including gastroesophageal junction adenocarcinoma. METHODS: This open-label, single arm, multicenter, phase 2 trial was conducted to assess first-line cetuximab plus S-1 plus cisplatin for advanced gastric cancer. A total of 40 patients from 10 centers were enrolled. Cetuximab was administered weekly, with the initial infusion at 400 mg/m2 and then 250 mg/m2 each subsequent week. S-1 plus cisplatin chemotherapy was concomitantly conducted in a 5-week cycle: S-1 (40-60 mg, adjusted for body surface area) was given twice daily for 3 consecutive weeks, followed by a 2-week rest period, and cisplatin (60 mg/m2) was given on day 8 of each cycle for a maximum of 8 cycles. Treatment continued until the occurrence of radiographically confirmed progressive disease, unacceptable toxicity or withdrawal of consent. The primary endpoint was the best overall response. Secondary endpoints included progression-free survival and safety. RESULTS: A total of 40 patients were evaluable. One patient (2.5%) had a complete response; 15 patients (37.5%) had a partial response. The observed overall response rate according to the independent review committee was 40.0% (95% confidence interval, 24.9-56.7; P = 0.7043 [one-sided null hypothesis: overall response rate ≤ 43%]); median PFS was 5.6 months (95% confidence intervals, 4.2-8.3). No adverse events leading to death were reported during the study, and no specific safety concerns were observed. CONCLUSIONS: Overall, the addition of cetuximab to S-1 plus cisplatin was well tolerated in patients with advanced gastric cancer but provided no additional clinical benefit in this study. ClinicalTrials.gov identifier: NCT01388790.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/administration & dosage , Cisplatin/administration & dosage , Oxonic Acid/administration & dosage , Stomach Neoplasms/drug therapy , Tegafur/administration & dosage , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Cetuximab/adverse effects , Cetuximab/therapeutic use , Cisplatin/adverse effects , Drug Administration Schedule , Drug Combinations , Female , Humans , Japan/epidemiology , Male , Middle Aged , Oxonic Acid/adverse effects , Remission Induction , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tegafur/adverse effects , Treatment Outcome
7.
BMC Cancer ; 20(1): 336, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32312286

ABSTRACT

BACKGROUND: The standard treatment for patients with unresectable locally advanced esophageal squamous cell carcinoma (ESCC) is definitive chemoradiotherapy (CRT) using 5-FU plus cisplatin. However, complete response (CR) rates are low at 11-25%, resulting in 9-10 months of median overall survival (OS). An improved therapeutic efficacy by combining immunotherapy with radiation has been reported in patients with locally advanced non-small cell lung cancer. The results using ESCC cell lines suggest sequential treatment with anti-PD-L1 agents soon after completion of CRT is the most effective combination. METHODS: TENERGY trial is a multicenter, phase II, proof-of-concept study to assess the efficacy and safety of atezolizumab following definitive CRT in patients with locally advanced ESCC. The main inclusion criteria are unresectable locally advanced ESCC without distant metastasis, completion of 60 Gy of radiation plus two concomitant cycles of chemotherapy (cisplatin 70 mg/m2 on day 1 and 5-FU 700 mg/m2 on days 1-4, every 28 days), and adequate organ function. Within 6 weeks after CRT, participants will start taking 1200 mg of atezolizumab every three weeks and continue until 12 months or disease progression. The primary endpoint is the confirmed CR rate by the investigator's assessment. Secondary endpoints include overall response rate, progression-free survival (PFS), OS, adverse events, and confirmed CR rate by central assessment. We will enroll 50 patients (40 with primary locally advanced ESCC and 10 with postoperative locoregionally recurrent ESCC). We will obtain biopsies from the primary site and will collect blood at 3 time points (before CRT, after CRT, and four weeks after the start of atezolizumab) for an exploratory biomarker study. We will analyze the phenotype of immune-competent cells, neoantigens, tumor mutational burden, PD-L1 status, and Human Leukocyte Antigen haplotyping. DISCUSSION: The synergistic efficacies of the sequential combination of CRT and atezolizumab should improve the CR rate, resulting in survival improvement for patients with unresectable locally advanced ESCC. Because CRT is a standard treatment option for patients with early stage to locally advanced ESCC, the sequential combination of CRT and atezolizumab has the potential to change the standard ESCC treatments. TRIAL REGISTRATION: UMIN000034373, 10/04/2018 and EPOC1802.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/mortality , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/therapy , Neoadjuvant Therapy/mortality , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Cisplatin/administration & dosage , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Survival Rate , Young Adult
8.
ESMO Open ; 5(1): e000624, 2020.
Article in English | MEDLINE | ID: mdl-33551068

ABSTRACT

BACKGROUND: While the BRAF V600E mutation occurs in 5%-15% of metastatic colorectal cancer (mCRC), BRAF non-V600E mutations were recently reported to range from 1.6% to 5.1%. We have previously reported that BRAF non-V600E mutations could have a negative impact on efficacy outcomes as well as BRAF V600E mutation for antiepidermal growth factor receptor (EGFR) antibody treatment for pretreated patients with mCRC. Recently, simultaneous inhibitions of mitogen-activated protein kinase kinase (MEK), BRAF and EGFR exhibited relevant antitumour activities in patients with BRAF V600E mutant and also in BRAF non-V600E mutant but only in the preclinical model. TRIAL DESIGN: The BIG BANG (study is a multicentre, phase II study to assess the efficacy, safety and proof of concept of the combinations of binimetinib+encorafenib+cetuximab in patients with BRAF non-V600E mutated mCRC, identified by either tumour tissue (tumour tissue group) or blood samples (liquid biopsy group). Key eligibility criteria include Eastern Cooperative Oncology Group Performance Status of ≤1, mCRC with BRAF non-V600E mutant and RAS wild type, refractory or intolerant to at least one fluoropyrimidine-based regimen and no prior history of regorafenib, and no prior history of anti-EGFR antibody treatment (primary analysis cohort and liquid biopsy cohort) or refractory to prior anti-EGFR antibody treatment in patients with class 3 BRAF mutations (anti-EGFR antibody refractory class three cohort). Enrolled patients receive binimetinib (45 mg, two times per day), encorafenib (300 mg, once a day) and cetuximab (initially 400 mg/m2 and subsequently 250 mg/m2, once per week). The primary endpoint is the confirmed objective response rate in the primary analysis cohort. TRIAL REGISTRATION NUMBERS: UMIN000031857 and 000031860.


Subject(s)
Colorectal Neoplasms , Adult , Antineoplastic Combined Chemotherapy Protocols , Benzimidazoles , Carbamates , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Female , HIV Infections , Humans , Male , Proto-Oncogene Proteins B-raf , Stroke Volume , Sulfonamides , Ventricular Function, Left , Young Adult
9.
ESMO Open ; 4(6): e000590, 2019.
Article in English | MEDLINE | ID: mdl-31798981

ABSTRACT

Background: BRAF V600E mutations are associated with aggressive biology and limited response to standard chemotherapy, especially during second-line and beyond therapies. BRAF V600E mutant and wild-type colorectal cancers (CRCs) differ in their expression profiles, and preclinical evidence suggests that microtubule inhibitors have an antitumour effect on xenograft models of BRAF V600E mutant CRCs. Eribulin has the best growth inhibitory activity in vitro of the microtubule inhibitors. Also, we have evidenced a hint of activity for patients with BRAF V600E mutant metastatic CRC (mCRC) with tumour shrinkage following eribulin treatment. Trial design: The BRAVERY study is a multicentre phase II study to evaluate the efficacy and safety of eribulin in patients with BRAF V600E mutant mCRC detected in either tumour tissues (primary analysis part) or circulating tumour DNA assays (liquid biopsy part). Key eligibility criteria are refractoriness and intolerance to at least one regimen (including irinotecan or oxaliplatin) containing fluoropyrimidine and Eastern Cooperative Oncology Group performance status of 0-1. Eribulin is to be administered intravenously at a dose of 1.4 mg/m2 on days 1 and 8 and repeated every 21 days. The primary endpoint is the confirmed objective response rate (ORR) by investigator's assessment. We calculated the sample size of the primary analysis part at 27 patients using a two-stage design with 25% ORR deemed promising and 5% unacceptable (one-sided α, 0.05; ß, 0.1). Secondary endpoints include disease control rate, progression-free survival, overall survival and adverse events. Moreover, we will collect pretreated tissue and serial blood samples for biomarker analyses, focusing on gene expression associated with BRAF mutant-like CRC to find predictive markers and acquired gene alterations to detect resistance mechanisms to eribulin. We initiated patient enrolment in March 2018, completed the primary analysis on May 2019, and are currently continuing with the liquid biopsy part. Trial registration number: UMIN000031221 and 000031552.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , Furans/administration & dosage , Ketones/administration & dosage , Research Design , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/blood , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Clinical Trials, Phase II as Topic , Colorectal Neoplasms/blood , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Drug Administration Schedule , Drug Resistance, Neoplasm/genetics , Female , Furans/adverse effects , Humans , Infusions, Intravenous , Irinotecan/pharmacology , Irinotecan/therapeutic use , Ketones/adverse effects , Liquid Biopsy/methods , Male , Multicenter Studies as Topic , Mutation , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Progression-Free Survival , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics
10.
Cancer Sci ; 110(9): 2875-2883, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31254422

ABSTRACT

Previous studies have shown sex-related differences in the incidence of adverse events following treatment with fluoropyrimidines, however the mechanism of this difference is unknown. We examined sex-related differences in the safety of S-1 plus oxaliplatin (SOX) and S-1 plus cisplatin (CS) in 663 metastatic gastric cancer patients taking part in a phase III study. The incidences of leukopenia (odds ratio [OR] 1.9; P = .015), neutropenia (OR 2.2; P = .002), nausea (OR 2.0; P = .009), and vomiting (OR 2.8; P < .001) were increased in women versus men treated with SOX, while vomiting (OR 2.9; P < .001) and stomatitis (OR 1.8; P = .043) were increased in women versus men treated with CS. In contrast, male patients treated with CS experienced thrombocytopenia more often (OR 0.51; P = .009). The mean relative dose intensity of S-1 in SOX was 75.4% in women and 81.4% in men (P = .032). No difference in efficacy was observed between women and men undergoing either regimen. Sex-related differences in adverse reactions during SOX and CS treatment were confirmed in this phase III study. Further translational research studies are warranted to pursue the cause of this difference.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/adverse effects , Oxaliplatin/adverse effects , Oxonic Acid/adverse effects , Stomach Neoplasms/drug therapy , Tegafur/adverse effects , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cisplatin/administration & dosage , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Combinations , Female , Humans , Incidence , Male , Middle Aged , Nausea/chemically induced , Nausea/epidemiology , Neutropenia/chemically induced , Neutropenia/epidemiology , Oxaliplatin/administration & dosage , Oxonic Acid/administration & dosage , Sex Factors , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Stomatitis/chemically induced , Stomatitis/epidemiology , Tegafur/administration & dosage , Thrombocytopenia/chemically induced , Thrombocytopenia/epidemiology , Vomiting/chemically induced , Vomiting/epidemiology
11.
Gastric Cancer ; 20(2): 332-340, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26956689

ABSTRACT

BACKGROUND: Adjuvant chemotherapy with XELOX (capecitabine plus oxaliplatin) has been shown to be beneficial following resection of gastric cancer in South Korean, Chinese, and Taiwanese patients. This phase II study (J-CLASSIC-PII) was undertaken to evaluate the feasibility of XELOX in Japanese patients with resected gastric cancer. METHODS: Patients with stage II or III gastric cancer who underwent curative D2 gastrectomy received adjuvant XELOX (eight 3-week cycles of oral capecitabine, 1000 mg/m2 twice daily on days 1-14, plus intravenous oxaliplatin 130 mg/m2 on day 1). The primary endpoint was dose intensity. Secondary endpoints were safety, proportion of patients completing treatment, and 1-year disease-free survival (DFS) rate. RESULTS: One hundred patients were enrolled, 76 of whom completed the study as planned. The mean dose intensity was 67.2 % (95 % CI, 61.9-72.5 %) for capecitabine and 73.4 % (95 % CI, 68.4-78.4 %) for oxaliplatin, which were higher than the predefined age-adjusted threshold values of 63.4 % and 69.4 %, respectively, and the study therefore met its primary endpoint. The 1-year DFS rate was 86 % (95 % CI, 77-91 %). No new safety signals were identified. CONCLUSIONS: The feasibility of adjuvant XELOX in Japanese patients with resected gastric cancer is similar to that observed in South Korean, Chinese, and Taiwanese patients in the Capecitabine and Oxaliplatin Adjuvant Study in Stomach Cancer (CLASSIC) study. Based on findings from this study and the CLASSIC study, the XELOX regimen can be considered an adjuvant treatment option for Japanese gastric cancer patients who have undergone curative resection.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Gastrectomy , Neoplasm Recurrence, Local/drug therapy , Stomach Neoplasms/drug therapy , Adult , Aged , Capecitabine , Chemotherapy, Adjuvant , Combined Modality Therapy , Deoxycytidine/therapeutic use , Female , Fluorouracil/therapeutic use , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Oxaloacetates , Prognosis , Stomach Neoplasms/pathology , Survival Rate
12.
Cancer Chemother Pharmacol ; 79(1): 89-98, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27915408

ABSTRACT

PURPOSE: To evaluate the safety, tolerability, pharmacokinetics, and efficacy of the intravenously administered pan-PI3K inhibitor copanlisib in Japanese patients with advanced or refractory solid tumors. METHODS: A Phase I open-label study in Japanese patients with advanced or refractory solid tumors was carried out. Patients received a single intravenous dose of either copanlisib 0.4 mg/kg or copanlisib 0.8 mg/kg, dosed intermittently on days 1, 8, and 15 of a 28-day cycle. Safety was monitored throughout the study. Plasma copanlisib levels were measured for pharmacokinetic analysis. RESULTS: Ten patients were enrolled and treated; three received copanlisib 0.4 mg/kg and seven received copanlisib 0.8 mg/kg. Overall, median duration of treatment was 6.2 weeks. No patients treated at 0.4 mg/kg experienced a dose-limiting toxicity, and the maximum tolerated dose in Japanese patients was determined to be 0.8 mg/kg. Adverse events were recorded in all ten patients; the most common were hyperglycemia, hypertension, and constipation. Copanlisib pharmacokinetic exposures displayed near dose-proportionality, with no accumulation. No patients achieved a complete or partial response, and disease control rate was 40.0%. CONCLUSIONS: Copanlisib was well tolerated in Japanese patients with advanced or refractory solid tumors, and the maximum tolerated dose was determined to be 0.8 mg/kg. Copanlisib demonstrated near dose-proportional pharmacokinetics and preliminary disease control, warranting further investigation. CLINICAL TRIAL REGISTRATION NUMBER: NCT01404390.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Aged , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Quinazolines/adverse effects , Quinazolines/pharmacokinetics
13.
Cancer Sci ; 107(5): 659-65, 2016 May.
Article in English | MEDLINE | ID: mdl-26918279

ABSTRACT

TAS-102, a novel oral antitumor agent, consists of trifluridine and tipiracil hydrochloride (molar ratio, 1:0.5). We investigated the effects of food on trifluridine and tipiracil hydrochloride. The efficacy and safety of TAS-102 were evaluated in patients with advanced solid tumors. We analyzed drug pharmacokinetics using a randomized, single-dose, two-treatment (fed versus fasting), two-period, two-sequence cross-over design, followed by repeated administration. Patients were given single doses of TAS-102 (35 mg/m(2) ) in the pharmacokinetic phase and received twice-daily doses of TAS-102 in 28-day cycles in the repeated administration phase for evaluating efficacy and safety. Food showed no effect on the area under the curve from 0 to 12 h or 0 h-infinity values of trifluridine following administration of TAS-102 under fasting and fed conditions, whereas those of tipiracil hydrochloride decreased by approximately 40%. Maximum concentrations of both drugs decreased by approximately 40%, indicating that food influenced the absorption and bioavailability of trifluridine and tipiracil hydrochloride, respectively. During the repeated administration, stable disease was observed in nine patients with rectal, small-cell lung, breast, thymic, duodenal, and prostate cancers. Major adverse events were neutropenia, leukopenia, anemia, and nausea. Postprandial administration was optimal for TAS-102 because trifluridine's area under the curve was not changed by food, indicating that its clinical efficacy would not be affected. Additionally, postprandial administration was reasonable because the maximum concentration of trifluridine decreased in neutrophils, which correlated with previous studies. These results suggest that TAS-102 would be an effective treatment for small-cell lung, thymic, and colorectal cancers. This trial is registered with the Japan Pharmaceutical Information Center (no. JapicCTI-111482).


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Food-Drug Interactions , Neoplasms/drug therapy , Neoplasms/metabolism , Trifluridine/pharmacology , Trifluridine/pharmacokinetics , Uracil/analogs & derivatives , Aged , Anemia/chemically induced , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Area Under Curve , Asian People , Biological Availability , Cross-Over Studies , Drug Combinations , Fasting , Female , Humans , Japan , Leukopenia/chemically induced , Male , Middle Aged , Nausea/chemically induced , Neoplasms/pathology , Neutropenia/chemically induced , Pyrrolidines/adverse effects , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Thymine/adverse effects , Thymine/pharmacokinetics , Thymine/pharmacology , Thymine/therapeutic use , Trifluridine/adverse effects , Trifluridine/therapeutic use , Uracil/adverse effects , Uracil/pharmacokinetics , Uracil/pharmacology , Uracil/therapeutic use
14.
Gan To Kagaku Ryoho ; 43(1): 65-8, 2016 Jan.
Article in Japanese | MEDLINE | ID: mdl-26809527

ABSTRACT

Prolonged prothrombin time is observed in patients taking warfarin (WF) with a fluoropyrimidine, such as S-1. When WF is combined with S-1, the prothrombin time-international normalized ratio (PT-INR) and dose adjustment of WF should be closely monitored. To date, no clinical data have been reported in terms of the relation between temporal variation of PT-INR and its therapeutic range. In this study, we retrospectively collected patients' clinical data including PT-INR. We identified 21 patients receiving WF therapy before the start of S-1 treatment. Patient characteristics were male/female: 18/3, median age: 69 (range 48-81) years old, cancer of gastric/lung/pancreatic/other: 8/5/4/4, and history of deep vein thrombosis (DVT)/atrial fibrillation (AF)/cerebral infarction (CI)/other: 11/6/2/2. The PT-INR of 16 patients exceeded normal upper limits after taking S-1 with WF. The median time to exceed the PT-INR upper therapeutic range is 25 (range 3-77) days. Patients receiving WF anticoagulant therapy concomitant with S-1 should have their PT-INR closely monitored and WF doses adjusted accordingly.


Subject(s)
Anticoagulants/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Neoplasms/drug therapy , Oxonic Acid/therapeutic use , Tegafur/therapeutic use , Warfarin/therapeutic use , Aged , Aged, 80 and over , Drug Combinations , Drug Interactions , Female , Humans , Male , Middle Aged , Prothrombin Time , Retrospective Studies
15.
Minerva Urol Nefrol ; 68(5): 437-43, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26554732

ABSTRACT

BACKGROUND: The aim of this study was to determine risk factors for survival after retrograde placement of ureteral stents and develop a prognostic model for advanced gastrointestinal tract (GIT: esophagus, stomach, colon and rectum) cancer patients. METHODS: We examined the clinical records of 122 patients who underwent retrograde placement of a ureteral stent against malignant extrinsic ureteral obstruction. A prediction model for survival after stenting was developed. We compared its clinical usefulness with our previous model based on the results from nephrostomy cases by decision curve analysis. RESULTS: Median follow-up period was 201 days (8-1490) and 97 deaths occurred. The 1-year survival rate in this cohort was 29%. Based on multivariate analysis, primary site of colon origin, absence of retroperitoneal lymph node metastasis and serum albumin >3g/dL were significantly associated with a prolonged survival time. To develop a prognostic model, we divided the patients into 3 risk groups of favorable: 0-1 factors (N.=53), intermediate: 2 risk factors (N.=54), and poor: 3 risk factors (N.=15). There were significant differences in the survival profiles of these 3 risk groups (P<0.0001). Decision curve analyses revealed that the current model has a superior net benefit than our previous model for most of the examined probabilities. CONCLUSIONS: We have developed a novel prognostic model for GIT cancer patients who were treated with retrograde placement of a ureteral stent. The current model should help urologists and medical oncologists to predict survival in cases of malignant extrinsic ureteral obstruction.


Subject(s)
Clinical Decision-Making , Gastrointestinal Neoplasms/surgery , Models, Theoretical , Stents , Ureter/surgery , Ureteral Obstruction/mortality , Ureteral Obstruction/surgery , Adult , Aged , Aged, 80 and over , Female , Gastrointestinal Neoplasms/complications , Gastrointestinal Neoplasms/mortality , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Risk Factors , Survival Rate , Ureteral Obstruction/etiology , Urologic Surgical Procedures/methods
16.
Gastric Cancer ; 19(1): 183-91, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25682441

ABSTRACT

BACKGROUND: This study was conducted to investigate whether human epidermal growth factor receptor 2 (HER2) status, epidermal growth factor receptor (EGFR) status, and c-MET status are independent prognostic factors for advanced gastric cancer patients who received standard chemotherapy. METHOD: Unresectable or recurrent gastric or gastroesophageal junction cancer patients with histologically confirmed adenocarcinoma treated with S-1 plus cisplatin as first-line chemotherapy were eligible. Formalin-fixed paraffin-embedded tumor samples were examined for HER2, EGFR, and c-MET status using immunohistochemistry (IHC). Additionally, gene amplification was examined using fluorescent in situ hybridization (FISH) for HER2. Positivity was defined as an IHC score of 3+ or an IHC score of 2+/FISH positive for HER2, and an IHC score of 2+ or 3+ for both EGFR and c-MET. RESULTS: Of the 293 patients from nine institutions, 43 (15%) were HER2 positive, 79 (27%) were EGFR positive, and 120 (41%) were c-MET positive. Ten patients (3%) showed positive co-expression of HER2, EGFR, and c-MET. After a median follow-up time of 58.4 months with 280 deaths, there was no significant difference in overall survival (OS) in terms of HER2 and EGFR status. However, there was a significant difference in OS between c-MET-positive and c-MET-negative patients [median, 11.9 months vs 14.2 months; hazard ratio, 1.31 (95% confidence interval, 1.03-1.67); log-rank P = 0.024]. Multivariate analysis also showed that c-MET positivity was still a prognostic factor for OS [hazard ratio, 1.30 (95% confidence interval, 1.02-1.67); P = 0.037]. CONCLUSIONS: The study suggested that c-MET-positive status had poor prognostic value. These data could be used as the basis for future clinical trials for targeting agents for advanced gastric cancer patients.


Subject(s)
ErbB Receptors/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cisplatin/administration & dosage , Drug Combinations , ErbB Receptors/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Oxonic Acid/administration & dosage , Prognosis , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/genetics , Retrospective Studies , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Tegafur/administration & dosage
17.
Cancer Sci ; 106(10): 1414-20, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26250827

ABSTRACT

We carried out a phase I/II trial of chemoradiotherapy concurrent with S-1 and cisplatin to determine the maximum tolerated dose and recommended dose and to evaluate the efficacy and safety of this treatment in patients with esophageal carcinoma. Thoracic esophageal cancer patients with clinical stage II/III disease, excluding T4, were eligible. Chemotherapy consisted of S-1 at a dose of 60-80 mg/m(2) /day on days 1-14, and cisplatin at 75 mg/m(2) on day 1, repeated twice every 4 weeks. Single daily radiation of 50.4 Gy was given in 28 fractions concurrently starting on day 1. Patients achieving an objective response after chemoradiotherapy underwent two additional cycles of chemotherapy. Patient accrual was terminated early due to slow enrolment after 44 patients were accrued. In the phase I part, two of six patients experienced dose-limiting toxicities at each level of S-1 (S-1 60 or 80 mg/m(2) /day). Considering treatment compliance, the recommended dose was determined to be S-1 60 mg/m(2) /day. The complete response rate, the primary endpoint of phase II, was 59.5% (22/37; 90% confidence interval, 44.6-73.1%; weighted threshold, 57.2%; P = 0.46 by the exact binomial test) on central review. In the phase II part, 3-year progression-free survival was 48.4%, with a 3-year overall survival of 61.9%. Grade 3 or 4 toxicity in phase II included leukopenia (57.9%), neutropenia (50%), hyponatremia (28.9%), anorexia (21.1%), anemia (18.4%), thrombocytopenia (18.4%), and febrile neutropenia (2.6%). No treatment-related deaths were observed. Although this combination showed acceptable toxicity and favorable 3-year survival, the study did not meet its primary endpoint. This trial was registered at the UMIN Clinical Trials Registry as UMIN000000710.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Squamous Cell/therapy , Cisplatin/therapeutic use , Esophageal Neoplasms/therapy , Oxonic Acid/therapeutic use , Pyridines/therapeutic use , Tegafur/therapeutic use , Adult , Aged , Antimetabolites, Antineoplastic/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Chemoradiotherapy/methods , Cisplatin/adverse effects , Disease-Free Survival , Drug Combinations , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/radiotherapy , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Oxonic Acid/adverse effects , Prospective Studies , Pyridines/adverse effects , Radiotherapy Dosage , Tegafur/adverse effects , Treatment Outcome , Young Adult
18.
Anticancer Drugs ; 26(10): 1043-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26288133

ABSTRACT

This phase I trial evaluated LY2603618, a selective inhibitor of the DNA damage checkpoint kinase 1, in combination with gemcitabine. Japanese patients with advanced solid tumors were enrolled. All patients received gemcitabine (1000 mg/m on days 1, 8, and 15 every 28 days) and either 170 mg (cohort 1) or 230 mg (cohort 2) of LY2603618. The primary objective was assessment of safety/tolerability. Pharmacokinetic/pharmacodynamic marker profiles were secondary objectives. Of the 17 patients enrolled, dose-limiting toxicities were observed in one patient in cohort 1 (n=7) and in two patients in cohort 2 (n=10). The most common grade 3 or more drug-related treatment-emergent adverse events were hematological. Three patients discontinued because of adverse events. Dose-dependent decreases in LY2603618 exposure were observed, but the LY2603618 pharmacokinetics at each dose were consistent within and between cycles and did not influence gemcitabine pharmacokinetics. Circulating plasma DNA decreased from baseline in all four patients who achieved a partial response. Administration of 170 or 230 mg of LY2603618 following a standard dose of gemcitabine showed acceptable safety and tolerability in Japanese patients with solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/metabolism , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Checkpoint Kinase 1 , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasms/pathology , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrazines/administration & dosage , Gemcitabine
19.
Invest New Drugs ; 33(5): 1068-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26163340

ABSTRACT

BACKGROUND: TAS-102 is a nucleoside antitumor agent consisting of trifluridine (FTD) and tipiracil hydrochloride (TPI). We investigated the recommended dose (RD) of TAS-102 plus irinotecan for metastatic colorectal cancer refractory to 5-fluorouracil (5-FU) and oxaliplatin. METHODS: This study was used a escalated dose of TAS-102 (40-70 mg/m(2)/day, for 5 days a week with 2 days rest for 2 weeks, followed by a 14-day rest) with a fixed dose of irinotecan (150 mg/m(2) on Days 1 and 15 of a 28-day schedule). The primary endpoints were determination of RD and assessment of safety. RESULTS: Ten patients were enrolled; 7 at the Level 1 (50 mg/m(2)/day) and 3 at the Level 2 (60 mg/m(2)/day). One patient at Level 1 was excluded from the analysis of dose-limiting toxicities (DLT) and efficacy. Five DLTs occurred in 3 patients; 1 patient at Level 1 (Grade 3 febrile neutropenia and Grade 4 neutropenia), and 2 patients at Level 2 (Grade 3 febrile neutropenia in two patients and Grade 4 neutropenia in one). Grade 3 or higher treatment-related adverse events were neutropenia (100 %), leukopenia (70 %), febrile neutropenia (30 %) and lymphopenia, anaemia (20 % each). 2 patients (22 %) achieved partial response with the duration of response were 112 and 799 days. CONCLUSION: The RD was determined to be 50 mg/m(2)/day of TAS-102 combined with 150 mg/m(2) of irinotecan although further investigation to explore optimal regimen is warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Area Under Curve , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Combinations , Female , Glucuronosyltransferase/genetics , Humans , Irinotecan , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis , Polymorphism, Genetic , Proto-Oncogene Proteins p21(ras)/genetics , Pyrrolidines , Thymine , Trifluridine/pharmacokinetics , Trifluridine/therapeutic use , Uracil/analogs & derivatives , Uracil/pharmacokinetics , Uracil/therapeutic use
20.
Cancer Chemother Pharmacol ; 76(2): 409-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26104654

ABSTRACT

PURPOSE: MK-2206 is an oral, highly selective inhibitor of AKT. The safety, tolerability, pharmacokinetics (PK), and anti-tumor activity of MK-2206 was evaluated in Japanese patients with advanced solid tumors. METHODS: Patients received a single oral dose of MK-2206 according to an every other day (QOD) dosing schedule or a once weekly (QW) dosing schedule in repeating 28-day treatment cycles, with a 7-day rest after only the first cycle. The dose-limiting toxicities (DLTs) were evaluated during Cycle 1. Full PK sampling was performed during Cycle 1. RESULTS: Twenty-four patients were treated at 45 mg (n = 3) or 60 mg (n = 9) QOD or at 135 mg (n = 3) or 200 mg (n = 9) QW. One patient experienced a DLT at 60 mg QOD, and three patients experienced DLTs at 200 mg QW. No DLTs were observed at 45 mg QOD or at 135 mg QW. The DLTs included mucosal inflammation, hyponatremia, face edema, erythema multiforme, and hyperglycemia. Common adverse events related to MK-2206 included rash, an elevated insulin c-peptide level, stomatitis, pyrexia, eosinophilia, leukopenia, and hyperglycemia. PK differences in MK-2206 exposure were observed between Japanese patients and non-Japanese patients. The higher exposure in Japanese patients was likely caused by the relatively lower weight of Japanese patients versus non-Japanese patients. No tumor responses were observed, but six patients exhibited stable disease lasting longer than 4 months. CONCLUSIONS: MK-2206 has an acceptable safety profile in Japanese patients with advanced solid tumors and warrants further investigation.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Administration, Oral , Adult , Aged , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Female , Gastrointestinal Neoplasms/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Leiomyosarcoma/drug therapy , Male , Middle Aged , Uterine Cervical Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...