Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Sci ; 115(2): 401-411, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38041233

ABSTRACT

Desmoid tumors (DTs), also called desmoid-type fibromatoses, are locally aggressive tumors of mesenchymal origin. In the present study, we developed a novel mouse model of DTs by inducing a local mutation in the Ctnnb1 gene, encoding ß-catenin in PDGFRA-positive stromal cells, by subcutaneous injection of 4-hydroxy-tamoxifen. Tumors in this model resembled histologically clinical samples from DT patients and showed strong phosphorylation of nuclear SMAD2. Knockout of SMAD4 in the model significantly suppressed tumor growth. Proteomic analysis revealed that SMAD4 knockout reduced the level of Cysteine-and-Glycine-Rich Protein 2 (CSRP2) in DTs, and treatment of DT-derived cells with a TGF-ß receptor inhibitor reduced CSRP2 RNA levels. Knockdown of CSRP2 in DT cells significantly suppressed their proliferation. These results indicate that the TGF-ß/CSRP2 axis is a potential therapeutic target for DTs downstream of TGF-ß signaling.


Subject(s)
Fibromatosis, Aggressive , Animals , Humans , Mice , beta Catenin/genetics , beta Catenin/metabolism , Fibromatosis, Aggressive/genetics , Fibromatosis, Aggressive/pathology , LIM Domain Proteins/genetics , Mice, Knockout , Muscle Proteins/metabolism , Nuclear Proteins/genetics , Proteomics , Transforming Growth Factor beta/metabolism , Up-Regulation
2.
Gastroenterology ; 164(2): 272-288, 2023 02.
Article in English | MEDLINE | ID: mdl-36155191

ABSTRACT

BACKGROUND & AIMS: We investigate interrelationships between gut microbes, metabolites, and cytokines that characterize COVID-19 and its complications, and we validate the results with follow-up, the Japanese 4D (Disease, Drug, Diet, Daily Life) microbiome cohort, and non-Japanese data sets. METHODS: We performed shotgun metagenomic sequencing and metabolomics on stools and cytokine measurements on plasma from 112 hospitalized patients with SARS-CoV-2 infection and 112 non-COVID-19 control individuals matched by important confounders. RESULTS: Multiple correlations were found between COVID-19-related microbes (eg, oral microbes and short-chain fatty acid producers) and gut metabolites (eg, branched-chain and aromatic amino acids, short-chain fatty acids, carbohydrates, neurotransmitters, and vitamin B6). Both were also linked to inflammatory cytokine dynamics (eg, interferon γ, interferon λ3, interleukin 6, CXCL-9, and CXCL-10). Such interrelationships were detected highly in severe disease and pneumonia; moderately in the high D-dimer level, kidney dysfunction, and liver dysfunction groups; but rarely in the diarrhea group. We confirmed concordances of altered metabolites (eg, branched-chain amino acids, spermidine, putrescine, and vitamin B6) in COVID-19 with their corresponding microbial functional genes. Results in microbial and metabolomic alterations with severe disease from the cross-sectional data set were partly concordant with those from the follow-up data set. Microbial signatures for COVID-19 were distinct from diabetes, inflammatory bowel disease, and proton-pump inhibitors but overlapping for rheumatoid arthritis. Random forest classifier models using microbiomes can highly predict COVID-19 and severe disease. The microbial signatures for COVID-19 showed moderate concordance between Hong Kong and Japan. CONCLUSIONS: Multiomics analysis revealed multiple gut microbe-metabolite-cytokine interrelationships in COVID-19 and COVID-19related complications but few in gastrointestinal complications, suggesting microbiota-mediated immune responses distinct between the organ sites. Our results underscore the existence of a gut-lung axis in COVID-19.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/genetics , Cross-Sectional Studies , SARS-CoV-2 , Feces/chemistry , Immunity , Cytokines , Vitamin B 6/analysis
3.
Cancer Sci ; 114(4): 1710-1717, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36601953

ABSTRACT

Comprehensive cancer genome profiling (CGP) has been nationally reimbursed in Japan since June 2019. Less than 10% of the patients have been reported to undergo recommended treatment. Todai OncoPanel (TOP) is a dual DNA-RNA panel as well as a paired tumor-normal matched test. Two hundred patients underwent TOP as part of Advanced Medical Care B with approval from the Ministry of Health, Labour and Welfare between September 2018 and December 2019. Tests were carried out in patients with cancers without standard treatment or when patients had already undergone standard treatment. Data from DNA and RNA panels were analyzed in 198 and 191 patients, respectively. The percentage of patients who were given therapeutic or diagnostic recommendations was 61% (120/198). One hundred and four samples (53%) harbored gene alterations that were detected with the DNA panel and had potential treatment implications, and 14 samples (7%) had a high tumor mutational burden. Twenty-two samples (11.1%) harbored 30 fusion transcripts or MET exon 14 skipping that were detected by the RNA panel. Of those 30 transcripts, 6 had treatment implications and 4 had diagnostic implications. Thirteen patients (7%) were found to have pathogenic or likely pathogenic germline variants and genetic counseling was recommended. Overall, 12 patients (6%) received recommended treatment. In summary, patients benefited from both TOP DNA and RNA panels while following the same indication as the approved CGP tests. (UMIN000033647).


Subject(s)
Genomics , Neoplasms , Humans , Japan , Neoplasms/drug therapy , Neoplasms/genetics , Precision Medicine
4.
Gastroenterology ; 163(4): 1038-1052, 2022 10.
Article in English | MEDLINE | ID: mdl-35788347

ABSTRACT

BACKGROUND & AIMS: Medication is a major determinant of human gut microbiome structure, and its overuse increases the risks of morbidity and mortality. However, effects of certain commonly prescribed drugs and multiple medications on the gut microbiome are still underinvestigated. METHODS: We performed shotgun metagenomic analysis of fecal samples from 4198 individuals in the Japanese 4D (Disease, Drug, Diet, Daily life) microbiome project. A total of 759 drugs were profiled, and other metadata, such as anthropometrics, lifestyles, diets, physical activities, and diseases, were prospectively collected. Second fecal samples were collected from 243 individuals to assess the effects of drug initiation and discontinuation on the microbiome. RESULTS: We found that numerous drugs across different treatment categories influence the microbiome; more than 70% of the drugs we profiled had not been examined before. Individuals exposed to multiple drugs, polypharmacy, showed distinct gut microbiome structures harboring significantly more abundant upper gastrointestinal species and several nosocomial pathobionts due to additive drug effects. Polypharmacy was also associated with microbial functions, including the reduction of short-chain fatty acid metabolism and increased bacterial stress responses. Even nonantibiotic drugs were significantly correlated with an increased antimicrobial resistance potential through polypharmacy. Notably, a 2-time points dataset revealed the alteration and recovery of the microbiome in response to drug initiation and cessation, corroborating the observed drug-microbe associations in the cross-sectional cohort. CONCLUSION: Our large-scale metagenomics unravels extensive and disruptive impacts of individual and multiple drug exposures on the human gut microbiome, providing a drug-microbe catalog as a basis for a deeper understanding of the role of the microbiome in drug efficacy and toxicity.


Subject(s)
Anti-Infective Agents , Gastrointestinal Microbiome , Microbiota , Cross-Sectional Studies , Fatty Acids, Volatile/pharmacology , Feces/microbiology , Gastrointestinal Microbiome/physiology , Humans , Metagenomics
5.
Gastroenterology ; 163(1): 222-238, 2022 07.
Article in English | MEDLINE | ID: mdl-35398347

ABSTRACT

BACKGROUND & AIMS: To identify gut and oral metagenomic signatures that accurately predict pancreatic ductal carcinoma (PDAC) and to validate these signatures in independent cohorts. METHODS: We conducted a multinational study and performed shotgun metagenomic analysis of fecal and salivary samples collected from patients with treatment-naïve PDAC and non-PDAC controls in Japan, Spain, and Germany. Taxonomic and functional profiles of the microbiomes were characterized, and metagenomic classifiers to predict PDAC were constructed and validated in external datasets. RESULTS: Comparative metagenomics revealed dysbiosis of both the gut and oral microbiomes and identified 30 gut and 18 oral species significantly associated with PDAC in the Japanese cohort. These microbial signatures achieved high area under the curve values of 0.78 to 0.82. The prediction model trained on the Japanese gut microbiome also had high predictive ability in Spanish and German cohorts, with respective area under the curve values of 0.74 and 0.83, validating its high confidence and versatility for PDAC prediction. Significant enrichments of Streptococcus and Veillonella spp and a depletion of Faecalibacterium prausnitzii were common gut signatures for PDAC in all the 3 cohorts. Prospective follow-up data revealed that patients with certain gut and oral microbial species were at higher risk of PDAC-related mortality. Finally, 58 bacteriophages that could infect microbial species consistently enriched in patients with PDAC across the 3 countries were identified. CONCLUSIONS: Metagenomics targeting the gut and oral microbiomes can provide a powerful source of biomarkers for identifying individuals with PDAC and their prognoses. The identification of shared gut microbial signatures for PDAC in Asian and European cohorts indicates the presence of robust and global gut microbial biomarkers.


Subject(s)
Metagenomics , Pancreatic Neoplasms , Dysbiosis/microbiology , Feces/microbiology , Humans , Metagenome , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Prospective Studies , Pancreatic Neoplasms
6.
Oncologist ; 27(10): e774-e782, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35946841

ABSTRACT

BACKGROUND: Gemcitabine plus nab-paclitaxel (GnP) has been a standard treatment for unresectable pancreatic cancer (uPC); however, the current treatment status and usefulness in older adults with uPC remain unclear. Therefore, we aimed to investigate the patient background and compare the efficacy and safety of GnP versus other treatments in older adults with uPC. PATIENTS AND METHODS: In this prospective observational study, we enrolled 233 eligible patients aged ≥76 years with pathologically proven, clinically uPC, and no history of chemotherapy from 55 Japanese centers during September 2018-September 2019. The main endpoints were overall survival (OS), progression-free survival (PFS), and safety. Geriatric assessments were performed upon registration and after 3 months. To adjust for confounders, we conducted propensity score-matched analyses. RESULTS: GnP, gemcitabine alone (Gem), best supportive care, and other therapies were administered to 116, 72, 16, and 29 patients, respectively. In the propensity score-matched analysis, 42 patients each were selected from the GnP and Gem groups. The median OS was longer in the GnP group than in the Gem group (12.2 vs. 9.4 months; hazard ratio [HR], 0.65; 95% CI, 0.37-1.13). The median PFS was significantly longer in the GnP group than in the Gem group (9.2 vs. 3.7 months; HR, 0.38; 95% CI, 0.23-0.64). The incidence of severe adverse events was higher with GnP than with Gem; however, the difference was not significant. CONCLUSION: GnP is more efficacious than Gem in patients aged ≥76 years with uPC despite demonstrating a higher incidence of severe adverse events.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Pancreatic Neoplasms , Aged , Albumins/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Deoxycytidine/analogs & derivatives , Humans , Paclitaxel , Pancreatic Neoplasms/drug therapy , Treatment Outcome , Gemcitabine , Pancreatic Neoplasms
7.
Pancreatology ; 22(8): 1159-1166, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36150984

ABSTRACT

BACKGROUND/OBJECTIVES: Pancreatic adenosquamous carcinoma (PASC) is a rare variant of pancreatic ductal adenocarcinoma (PDAC). The usual treatment for metastatic or recurrent PASC is systemic chemotherapy in accordance with the PDAC treatment strategy. This study aimed to investigate the efficacy of chemotherapy, especially the benefit of recent combination therapies, in patients with metastatic or recurrent PASC. METHODS: We conducted a multicenter retrospective analysis of 116 patients with metastatic or recurrent PASC treated with first-line chemotherapy between April 2001 and December 2017 at 24 Japanese institutions. RESULTS: Combination chemotherapies included gemcitabine + nab-paclitaxel (GnP, n = 28), fluorouracil/leucovorin + irinotecan + oxaliplatin (FFX, n = 10), gemcitabine + S-1 (GS, n = 10), and others (n = 9). Monotherapies included gemcitabine (n = 51) and S-1 (n = 8). The median overall survival (OS) was 6.5, 7.3, and 4.3 months for the whole cohort, the combination therapy group, and the monotherapy group, respectively. Multivariate analysis indicated that combination therapy showed a better trend in OS than monotherapy (hazard ratio = 0.68; 95% confidence interval, 0.38-1.20). GnP or FFX were selected in 58.7% of patients after FFX was approved in Japan, and revealed a median OS, median progression-free survival, and objective response rate of 7.3 months, 2.8 months, and 26.9% in GnP and 7.2 months, 2.3 months, and 20.0% in FFX respectively. CONCLUSIONS: This study suggests that combination therapy may be more effective than monotherapy. GnP and FFX showed similar and clinically meaningful efficacy for patients with metastatic or recurrent PASC.


Subject(s)
Carcinoma, Adenosquamous , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Retrospective Studies , Carcinoma, Adenosquamous/drug therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms
8.
Ann Surg Oncol ; 27(2): 460-467, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31376034

ABSTRACT

BACKGROUND: A multicenter phase 2 trial analysed chemoselection with docetaxel plus 5-fluorouracil and cisplatin (DCF) induction chemotherapy (ICT) and subsequent conversion surgery (CS) for locally advanced unresectable esophageal cancer. This study presents updated 3-year analyses to further characterize the impact of DCF-ICT followed by CS. METHODS: Esophageal cancer patients with clinical T4 disease, unresectable supraclavicular lymph node metastasis, or both were eligible for this study. The treatment starts with DCF-ICT, followed by CS if the cancer is resectable, or by concurrent chemoradiation if it is not resectable. This updated analysis presents 3-year overall survival (OS), 3-year progression-free survival (PFS), and pattern of relapse. RESULTS: The median follow-up period for the patients surviving without death was 39.3 months. The estimated 1-year OS was 66.7%, and the lower limit of the 80% confidence interval (CI) was 54.6%. The estimated 3-year OS was 46.6% (95% CI 34.2-63.5%). The OS for the patients who underwent R0 resection (n = 19) was significantly longer than for those who did not (3-year OS: 71.4% vs. 30.1%). The estimated 1-year PFS was 50.6%, and the 3-year PFS was 39.6%. The PFS for R0 was significantly longer than for non-R0 (3-year PFS: 61.3% vs 25.0%). Recurrence or progression at the primary site was observed in 31% of the non-R0 group. The rate of distant metastasis did not differ significantly between the non-R0 and R0 groups (21% vs 16%). CONCLUSIONS: Long-term follow-up evaluation confirmed that DCF chemoselection aimed at CS is feasible and promising in terms of survival for patients with locally advanced esophageal cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy/mortality , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/mortality , Esophagectomy/mortality , Induction Chemotherapy/mortality , Neoplasm Recurrence, Local/mortality , Aged , Cisplatin/administration & dosage , Docetaxel/administration & dosage , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/therapy , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Prognosis , Survival Rate
9.
Pancreatology ; 20(7): 1421-1427, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32891532

ABSTRACT

BACKGROUND/OBJECTIVES: Pancreatic neuroendocrine carcinoma (PanNEC)-G3 often presents along with genetic abnormalities such as KRAS, RB1, and TP53 mutations. However, the association between these genetic findings and response to chemotherapy and prognosis has not been clarified. This study aimed to clarify the clinicopathological features of PanNEC-G3. METHODS: We performed a subgroup analysis of the Japanese PanNEN-G3 study (multicenter, retrospective study), which revealed that Rb loss and KRAS mutation were predictors of the response to platinum-based regimen in PanNEN-G3. We re-classified WHO grades of PanNENs using the 2017 WHO classification and then analyzed the clinicopathological features and prognostic factors in 49 patients with PanNEC-G3. RESULTS: The rates of Rb loss and KRAS mutation in PanNEC-G3 were 54.5% and 48.7%, respectively. Patients with Rb loss and/or KRAS mutation showed a higher response rate to first-line platinum-based regimen than those without Rb loss or KRAS mutation (object response rate 70.0% vs 33.3%, odds ratio 9.22; 95% CI 1.26-67.3, P = 0.029), but tended to have shorter overall survival rates than those without Rb loss or KRAS mutation (median 239 vs 473 days, hazard ratio 2.11; 95% CI 0.92-4.86, P = 0.077). CONCLUSIONS: Patients with PanNEC-G3 have varied clinical outcomes for platinum-based regimen. When grouped based on Rb loss and KRAS mutation, there seemed to be two groups with distinct prognoses and responses to the platinum-based regimen. PanNEC-G3 could, therefore, be classified into two distinct groups based on immunohistochemical and genetic findings.


Subject(s)
Carcinoma, Neuroendocrine/classification , Pancreatic Neoplasms/classification , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/genetics , Endoscopic Ultrasound-Guided Fine Needle Aspiration , Female , Humans , Japan , Male , Middle Aged , Neoplasm Grading , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Platinum Compounds/therapeutic use , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Retinoblastoma Binding Proteins/genetics , Retrospective Studies , Survival Analysis , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
10.
Neuroendocrinology ; 110(11-12): 988-993, 2020.
Article in English | MEDLINE | ID: mdl-31986515

ABSTRACT

BACKGROUND: Platinum-containing regimens are widely used as first-line chemotherapy for unresectable pancreatic neuroendocrine carcinoma (NEC), but second-line chemotherapies have yet to be established. OBJECTIVES: We evaluated the safety and efficacy of everolimus in patients with pancreatic NEC refractory or intolerant to platinum-containing chemotherapy. METHODS: This study was a prospective, multicenter, phase II trial in patients with pancreatic NEC after platinum-containing chemotherapy. Everolimus treatment was continued until disease progression or intolerable toxicity was observed. The primary endpoint was progression-free survival (PFS). RESULTS: Participants comprised 25 patients. Median age was 63 years, median PFS was 1.2 months (95% confidence interval [CI] 0.9-3.1 months), median overall survival was 7.5 months (95% CI 3.1-13.5 months), overall response rate was 0%, and disease control rate was 39.1%. Common grade 3/4 adverse events were hyperglycemia (20%), thrombocytopenia (16%), and anemia (16%). CONCLUSION: The efficacy of everolimus was limited in patients with unresectable pancreatic NEC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/drug therapy , Everolimus/pharmacology , Outcome Assessment, Health Care , Pancreatic Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Drug-Related Side Effects and Adverse Reactions , Everolimus/administration & dosage , Everolimus/adverse effects , Female , Humans , Male , Middle Aged , Platinum/therapeutic use , Progression-Free Survival , Prospective Studies
11.
Digestion ; 101(2): 174-183, 2020.
Article in English | MEDLINE | ID: mdl-30897577

ABSTRACT

BACKGROUND/AIM: We evaluated the efficacy of vonoprazan (VPZ), a novel potassium-competitive acid blocker, in patients with proton pump inhibitor (PPI)-refractory gastroesophageal reflux disease (GERD), exhibiting continued pathological esophageal acid exposure (EAE). METHODS: Despite ≥8 weeks of appropriate PPI therapy, patients with -persistent reflux symptoms and pathological EAE times (EAETs ≥4%) were invited to switch to VPZ treatment. After an 8-week-course of once-daily VPZ (20 mg), multichannel intraluminal impedance-pH (MII-pH) monitoring was repeated to compare gastric acid exposure times (GAETs), EAETs, and other reflux parameters relative to the baseline values. Before each MII-pH study, reflux symptom severities were scored using the Gastrointestinal Symptom Rating Scale; erosive esophagitis and fasting plasma gastrin levels were also assessed. RESULTS: From among the 124 patients undergoing MII-pH monitoring, 13 patients (median age, 69 years; females, 64%) were monitored at baseline (while on PPI therapy) and after VPZ therapy. The median GAET associated with VPZ treatment (23.8%) was less than that for PPI treatment (41.1%; p = 0.01), including both daytime and nighttime measurements. VPZ therapy resulted in better median EAET values (4.5%) than did PPI therapy (10.6%) during the 24-h monitoring period (p = 0.055). EAE normalization was achieved in 46% of VPZ-treated patients and was associated with complete gastric acid suppression (p = 0.005). After switching to VPZ, reflux symptoms (p < 0.01) and erosive esophagitis (p = 0.01) improved. CONCLUSION: In patients with PPI-refractory GERD, VPZ provides more potent gastric acid suppression, more effective EAE control, enhanced symptom improvement, and better esophagitis healing than PPIs.


Subject(s)
Esophagitis, Peptic/drug therapy , Gastroesophageal Reflux/drug therapy , Proton Pump Inhibitors/therapeutic use , Pyrroles/therapeutic use , Sulfonamides/therapeutic use , Aged , Aged, 80 and over , Drug Substitution , Esophageal pH Monitoring , Esophagitis, Peptic/etiology , Female , Gastroesophageal Reflux/complications , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome
12.
Proc Natl Acad Sci U S A ; 114(37): E7697-E7706, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28847964

ABSTRACT

Cancer cells alter their metabolism for the production of precursors of macromolecules. However, the control mechanisms underlying this reprogramming are poorly understood. Here we show that metabolic reprogramming of colorectal cancer is caused chiefly by aberrant MYC expression. Multiomics-based analyses of paired normal and tumor tissues from 275 patients with colorectal cancer revealed that metabolic alterations occur at the adenoma stage of carcinogenesis, in a manner not associated with specific gene mutations involved in colorectal carcinogenesis. MYC expression induced at least 215 metabolic reactions by changing the expression levels of 121 metabolic genes and 39 transporter genes. Further, MYC negatively regulated the expression of genes involved in mitochondrial biogenesis and maintenance but positively regulated genes involved in DNA and histone methylation. Knockdown of MYC in colorectal cancer cells reset the altered metabolism and suppressed cell growth. Moreover, inhibition of MYC target pyrimidine synthesis genes such as CAD, UMPS, and CTPS blocked cell growth, and thus are potential targets for colorectal cancer therapy.


Subject(s)
Adenoma/metabolism , Colorectal Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Adenoma/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , Disease Models, Animal , Female , Genes, myc , Humans , Male , Metabolomics/methods , Mice , Proto-Oncogene Proteins c-myc/genetics , Pyrimidines/biosynthesis , Transcriptome
13.
Cancer Sci ; 110(8): 2520-2528, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31215118

ABSTRACT

Iodothyronine deiodinase 2 (DIO2) converts the prohormone thyroxine (T4) to bioactive T3 in peripheral tissues and thereby regulates local thyroid hormone (TH) levels. Although epidemiologic studies suggest the contribution of TH to the progression of colorectal cancer (CRC), the role of DIO2 in CRC remains elusive. Here we show that Dio2 is highly expressed in intestinal polyps of ApcΔ716 mice, a mouse model of familial adenomatous polyposis and early stage sporadic CRC. Laser capture microdissection and in situ hybridization analysis show almost exclusive expression of Dio2 in the stroma of ApcΔ716 polyps in the proximity of the COX-2-positive areas. Treatment with iopanoic acid, a deiodinase inhibitor, or chemical thyroidectomy suppresses tumor formation in ApcΔ716 mice, accompanied by reduced tumor cell proliferation and angiogenesis. Dio2 expression in ApcΔ716 polyps is strongly suppressed by treatment with the COX-2 inhibitor meloxicam. Analysis of The Cancer Genome Atlas data shows upregulation of DIO2 in CRC clinical samples and a close association of its expression pattern with the stromal component, consistently with almost exclusive expression of DIO2 in the stroma of human CRC as revealed by in situ hybridization. These results indicate essential roles of stromal DIO2 and thyroid hormone signaling in promoting the growth of intestinal tumors.


Subject(s)
Cell Proliferation/physiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Iodide Peroxidase/metabolism , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/pathology , Animals , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Models, Animal , Humans , Intestinal Polyps/drug therapy , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Mice , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , Thyroid Hormones/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , Iodothyronine Deiodinase Type II
14.
Ann Surg Oncol ; 26(5): 1385-1393, 2019 May.
Article in English | MEDLINE | ID: mdl-30863939

ABSTRACT

BACKGROUND: The role of surgery in pancreatic neuroendocrine neoplasm grade 3 (pNEN-G3) treatment remains unclear. We aimed to clarify the role of surgery for pNEN-G3, which has recently been reclassified as pancreatic neuroendocrine tumor-G3 (pNET-G3) and pancreatic neuroendocrine carcinoma-G3 (pNEC-G3), with and without metastases, respectively. METHODS: We analyzed a subgroup of patients from the Japanese pancreatic NEC study, a Japanese multicenter case-series study of pNEN-G3. Pathologists subclassified 67 patients as having pNET-G3 or pNEC-G3 based on morphological features. We compared the overall survival (OS) rates among patients who were grouped according to whether they had undergone tumor-targeted surgery for tumors without (SwoM) or with (SwM) metastases, or non-surgical procedures (NS). RESULTS: Data from 21 patients with pNET-G3 (SwoM, n = 6; SwM, n = 5; NS, n = 10) and 46 patients with pNEC-G3 (SwoM, n = 8; SwM, n = 5; NS, n = 33) were analyzed. OS of patients with pNET-G3 was significantly longer after SwoM and SwM than with NS (p = 0.018 and p = 0.022). In contrast, OS did not significantly differ between either SwoM or SwM and NS (p = 0.093 and p = 0.489) among patients with pNEC-G3. CONCLUSION: The role of surgery should be considered separately for pNET-G3 and pNEC-G3. Although SwoM and SwM can be considered for pNET-G3, caution is advised before considering SwM and SwoM for pNEC-G3.


Subject(s)
Carcinoma, Neuroendocrine/mortality , Neuroendocrine Tumors/mortality , Pancreatectomy/mortality , Pancreatic Neoplasms/mortality , Adult , Aged , Aged, 80 and over , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/surgery , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/surgery , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Survival Rate
15.
Gan To Kagaku Ryoho ; 46(12): 1818-1822, 2019 Dec.
Article in Japanese | MEDLINE | ID: mdl-31879395

ABSTRACT

Cachexia is considered as a complex metabolic disease accompanied by systemic inflammation. However, we still do not understand the essential nature of the metabolic disorder associated with cachexia or the precise molecular mechanisms that drive cachexia. This reviewsummarizes the current knowledge on the pathogenesis of cancer cachexia obtained mainly from mouse models with emphasis on the findings that could reach the bedside in the future. Basic studies using animal models of cancer cachexia indicate that mediators such as pro-inflammatory cytokines and members of TGF-b superfamily disturb the cross-talks among metabolism-related organs including skeletal muscle, adipose tissue, liver, and CNS and thereby induce the collapse of metabolic homeostasis. The inhibitors of these mediators are currently under development for the treatment of cancer cachexia. Skeletal muscle atrophy is a key feature of cancer cachexia and is induced by enhanced proteolysis via ubiquitin-proteasome system and autophagy-lysosome system, as well as by decreased protein synthesis and increased fatty acid oxidation. Adipose tissue atrophy due to excessive lipolysis is another common feature of cancer cachexia, and the involvement of the browning of white adipose tissue and of the increased energy expenditure associated with the futile cycle of lipolysis/lipogenesis is suggested. The liver of cachectic tumor-bearing mice shows increased gluconeogenesis which leads to energy expenditure via futile cycle, and also develops steatosis due to decreased triglyceride usage. In the CNS, inflammation in the hypothalamus induces anorexia and excessive peripheral energy expenditure. Cachectic animals also showresistance to the appetite-promoting effects of ghrelin. We hope that cancer cachexia will gain better awareness in the near future, leading to the growth and progress of the research field, and that the elucidation of its pathogenesis will contribute to the development of novel preventive/therapeutics strategies.


Subject(s)
Cachexia , Neoplasms , Adipose Tissue , Animals , Anorexia , Cachexia/etiology , Cytokines , Mice , Muscle, Skeletal , Neoplasms/complications
16.
Cancer Sci ; 109(8): 2549-2557, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29856900

ABSTRACT

We conducted a multicenter retrospective analysis to evaluate the efficacy of systemic chemotherapy for unresectable combined hepatocellular and cholangiocarcinoma. We enrolled 36 patients with pathologically proven, unresectable combined hepatocellular and cholangiocarcinoma treated with systemic chemotherapy. The log-rank test determined the significance of each prognostic factor. Elevated alpha-fetoprotein, carcinoembryonic antigen and carbohydrate antigen 19-9 levels were observed in 58.3%, 16.7% and 38.9% of patients, respectively. First-line chemotherapy included platinum-containing regimens consisting of gemcitabine/cisplatin (n = 12) and fluorouracil/cisplatin (n = 11), sorafenib (n = 5) and others (n = 8). The median overall and progression-free survival times were 8.9 and 2.8 months, respectively, with an overall response rate of 5.6%. Prognostic factors associated with negative outcomes included poor performance status, no prior primary tumor resection, a Child-Pugh class of B, and elevated carcinoembryonic antigen levels with a hazard ratio of 2.25, 2.48, 3.25 and 2.84 by univariate analysis, respectively. The median overall survival times of the gemcitabine/cisplatin, fluorouracil/cisplatin, sorafenib and other groups were 11.9, 10.2, 3.5 and 8.1 months, respectively. Multivariate analysis revealed that the overall survival of patients within the sorafenib monotherapy group was poor compared with platinum-containing regimens (HR: 15.83 [95% CI: 2.25-111.43], P = .006). All 7 patients in the sorafenib group had progressive disease, including 2 patients with second-line therapy. In conclusion, the platinum-containing regimens such as gemcitabine/cisplatin were associated with more favorable outcomes than sorafenib monotherapy for unresectable combined hepatocellular and cholangiocarcinoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bile Duct Neoplasms/drug therapy , Carcinoma, Hepatocellular/drug therapy , Cholangiocarcinoma/drug therapy , Liver Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Bile Ducts, Intrahepatic/drug effects , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Humans , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Retrospective Studies , Young Adult
17.
BMC Cancer ; 18(1): 993, 2018 Oct 19.
Article in English | MEDLINE | ID: mdl-30340559

ABSTRACT

BACKGROUND: Epithelioid hemangioendothelioma is an exceedingly rare sarcoma often occurring as an indolent angiocentric vascular tumor at various anatomic sites. Few reports have evaluated large case series of epithelioid hemangioendothelioma. METHODS: We conducted a retrospective analysis of the clinical data of 42 consecutive patients with epithelioid hemangioendothelioma who were pathologically diagnosed between 1990 and 2014 at 13 Japanese tertiary hospitals. We analyzed their clinical characteristics, tumor features and prognostic factors. RESULTS: The study included 22 men and 20 women, with a median age of 54 (range, 18-78) years. Pain was the most common symptom, occurring in 15 (68%) of the 22 symptomatic patients. The median maximum tumor diameter was 4.0 (range, 1.0-12.8) cm. The most commonly involved organs were the liver (81%), lungs (57%), and bones (12%). The overall survival rates were 79.5% at 1 year and 72.0% at 5 years. Substantially better survival was observed in asymptomatic patients than in symptomatic patients (P = 0.03), and better survival was also ovserved in patients with Ki-67 index ≤10% than in those with Ki-67 index > 10% (P = 0.04). By multivariate analysis, tumor size > 3.0 cm was associated with decreased survival (P = 0.049, hazard ratio 13.33). CONCLUSIONS: This study showed the clinical characteristics of Japanese patients with epithelioid hemangioendothelioma. Tumor size > 3.0 cm is an independent indicator of a poor prognosis in epithelioid hemangioendothelioma. The presence of symptoms at the time of diagnosis and high Ki-67 index implied poor survival.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/mortality , Hemangioendothelioma, Epithelioid/diagnosis , Hemangioendothelioma, Epithelioid/mortality , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/mortality , Adolescent , Adult , Aged , Female , Humans , Japan/epidemiology , Male , Middle Aged , Neoplasms, Vascular Tissue/diagnosis , Neoplasms, Vascular Tissue/mortality , Retrospective Studies , Survival Rate/trends , Young Adult
18.
Gastric Cancer ; 21(3): 439-445, 2018 May.
Article in English | MEDLINE | ID: mdl-28766263

ABSTRACT

BACKGROUND: A combination of S-1 and cisplatin is recognized as one of the standard first-line chemotherapy regimens for patients with advanced gastric cancer. However, demographic analyses of pivotal phase III studies have showed that only a minority of treated patients were aged 76 years or older. The purpose of this phase II study was to evaluate the safety and efficacy of combination therapy with S-1 and cisplatin in elderly patients with chemotherapy-naive advanced gastric cancer. METHODS: Patients aged 76 years or older received S-1 40 mg/m2 orally twice daily for 21 days and cisplatin 60 mg/m2 intravenously infused at day 8 of each 35-day cycle. Dose modification was performed according to creatinine clearance. The primary endpoint was overall survival (OS). Secondary endpoints included response rate, progression-free survival (PFS), time to treatment failure (TTF), and adverse events. RESULTS: A total of 40 patients were enrolled. Median OS was 12.3 months, PFS was 7.8 months, and TTF was 4.3 months. The response rate was 54%. The most common grade 3-4 adverse events were anorexia (25%), neutropenia (23%), hyponatremia (20%), anemia (18%), and febrile neutropenia (8%). No treatment-related death occurred. CONCLUSIONS: Combination chemotherapy with S-1 and cisplatin is an effective and well-tolerated regimen for elderly patients with advanced gastric cancer when the dose is adjusted according to renal function.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Stomach Neoplasms/drug therapy , Adenocarcinoma/mortality , Administration, Oral , Aged , Aged, 80 and over , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease-Free Survival , Drug Combinations , Female , Humans , Kaplan-Meier Estimate , Male , Oxonic Acid/administration & dosage , Oxonic Acid/adverse effects , Stomach Neoplasms/mortality , Tegafur/administration & dosage , Tegafur/adverse effects , Treatment Outcome
19.
Undersea Hyperb Med ; 45(1): 41-48, 2018.
Article in English | MEDLINE | ID: mdl-29571231

ABSTRACT

BACKGROUND: Decompression sickness (DCS) is a rare condition that is often difficult to diagnose in deep-sea divers. Because of this, prevention and early diagnosis are important. In this case-control study, we examined the risk and preventive factors associated with DCS. METHODS: Our original questionnaire survey was conducted among 269 recreational divers in Okinawa. Divers who were diagnosed with DCS by a physician (n = 94) were compared with healthy recreational divers (n = 175). The questionnaire consisted of 30 items and included a dive profile. Odds ratios and multiple logistic regression analysis were used to estimate the relative risk of DCS. RESULTS: Logistic regression analysis revealed the following risk factors for DCS: a past history of DCS, drinking alcohol the evening before diving, indicating decompression stops, cold exposure after the dive, and maximum depth. Preventive factors included hydration before the dive, deep stops, safety stops and using nitrox gas. The results were reliable according to the Hosmer-Lemeshow and omnibus tests. CONCLUSION: We identified certain risk factors, together with their relative risks, for DCS. These risk factors may facilitate prevention of DCS among Okinawa divers.


Subject(s)
Decompression Sickness/etiology , Decompression Sickness/prevention & control , Diving/adverse effects , Adult , Aged , Alcohol Drinking/adverse effects , Case-Control Studies , Cold Temperature/adverse effects , Confidence Intervals , Female , Health Surveys , Humans , Japan , Male , Middle Aged , Odds Ratio , Regression Analysis , Risk Factors , Young Adult
20.
Int J Clin Pharmacol Ther ; 55(5): 409-415, 2017 May.
Article in English | MEDLINE | ID: mdl-28157071

ABSTRACT

OBJECTIVE: Due to the occurrence of severe adverse drug reactions to regorafenib, a drug used in cancer therapy, the identification of a predictive marker(s) is needed to increase the therapeutic applicability of this compound. We therefore investigated whether polymorphisms in the ABCG2 and SLCO1B genes are associated with adverse drug reactions to regorafenib. METHODS: For these analyses, 37 Japanese cancer patients were treated with regorafenib, genotyped for polymorphisms in ABCG2 and SLCO1B, and evaluated for drug-related adverse drug reactions. RESULTS: There was no association between the ABCG2 421C>A variant and adverse drug reactions to regorafenib. After treatment, the incidences of increased aspartate aminotransferase (AST) and alanine aminotransferase (ALT) as well as increased total bilirubin (grade ≥ 2) were 8%, 4%, and 12%, and 42%, 25%, and 25% among SLCO1B1*1b carriers and non-carriers, respectively. There were no significant associations between elevated ALT and bilirubin and the SLCO1B1*1b allele. However, there were significantly lower incidences of increased AST (8% vs. 42%) and anemia (16% vs. 50%) in SLCO1B1*1b carriers than in non-carriers. CONCLUSIONS: The absence of SLCO1B1*1b allele appears to be associated with the development of adverse drug reactions to regorafenib; however, further studies involving larger test groups and other populations are needed to confirm these findings.
.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Anemia/genetics , Antineoplastic Agents/adverse effects , Chemical and Drug Induced Liver Injury/genetics , Liver-Specific Organic Anion Transporter 1/genetics , Neoplasm Proteins/genetics , Pharmacogenomic Variants , Phenylurea Compounds/adverse effects , Polymorphism, Genetic , Protein Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Adult , Aged , Aged, 80 and over , Anemia/blood , Anemia/chemically induced , Anemia/diagnosis , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/diagnosis , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Japan , Male , Middle Aged , Pharmacogenetics , Pharmacogenomic Testing/methods , Phenotype , Predictive Value of Tests , Prospective Studies , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL