Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Nature ; 615(7950): 168-174, 2023 03.
Article in English | MEDLINE | ID: mdl-36813961

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is expected to be the second most deadly cancer by 2040, owing to the high incidence of metastatic disease and limited responses to treatment1,2. Less than half of all patients respond to the primary treatment for PDAC, chemotherapy3,4, and genetic alterations alone cannot explain this5. Diet is an environmental factor that can influence the response to therapies, but its role in PDAC is unclear. Here, using shotgun metagenomic sequencing and metabolomic screening, we show that the microbiota-derived tryptophan metabolite indole-3-acetic acid (3-IAA) is enriched in patients who respond to treatment. Faecal microbiota transplantation, short-term dietary manipulation of tryptophan and oral 3-IAA administration increase the efficacy of chemotherapy in humanized gnotobiotic mouse models of PDAC. Using a combination of loss- and gain-of-function experiments, we show that the efficacy of 3-IAA and chemotherapy is licensed by neutrophil-derived myeloperoxidase. Myeloperoxidase oxidizes 3-IAA, which in combination with chemotherapy induces a downregulation of the reactive oxygen species (ROS)-degrading enzymes glutathione peroxidase 3 and glutathione peroxidase 7. All of this results in the accumulation of ROS and the downregulation of autophagy in cancer cells, which compromises their metabolic fitness and, ultimately, their proliferation. In humans, we observed a significant correlation between the levels of 3-IAA and the efficacy of therapy in two independent PDAC cohorts. In summary, we identify a microbiota-derived metabolite that has clinical implications in the treatment of PDAC, and provide a motivation for considering nutritional interventions during the treatment of patients with cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Microbiota , Pancreatic Neoplasms , Animals , Humans , Mice , Carcinoma, Pancreatic Ductal/diet therapy , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/microbiology , Glutathione Peroxidase/metabolism , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/microbiology , Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Tryptophan/metabolism , Tryptophan/pharmacology , Tryptophan/therapeutic use , Neutrophils/enzymology , Autophagy , Metagenome , Metabolomics , Fecal Microbiota Transplantation , Indoleacetic Acids/pharmacology , Indoleacetic Acids/therapeutic use , Disease Models, Animal , Germ-Free Life , Pancreatic Neoplasms
2.
Med ; 3(2): 119-136, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35425930

ABSTRACT

Background: Ketogenic diet is a potential means of augmenting cancer therapy. Here, we explore ketone body metabolism and its interplay with chemotherapy in pancreatic cancer. Methods: Metabolism and therapeutic responses of murine pancreatic cancer were studied using KPC primary tumors and tumor chunk allografts. Mice on standard high-carbohydrate diet or ketogenic diet were treated with cytotoxic chemotherapy (nab-paclitaxel, gemcitabine, cisplatin). Metabolic activity was monitored with metabolomics and isotope tracing, including 2H- and 13C-tracers, liquid chromatography-mass spectrometry, and imaging mass spectrometry. Findings: Ketone bodies are unidirectionally oxidized to make NADH. This stands in contrast to the carbohydrate-derived carboxylic acids lactate and pyruvate, which rapidly interconvert, buffering NADH/NAD. In murine pancreatic tumors, ketogenic diet decreases glucose's concentration and tricarboxylic acid cycle contribution, enhances 3-hydroxybutyrate's concentration and tricarboxylic acid contribution, and modestly elevates NADH, but does not impact tumor growth. In contrast, the combination of ketogenic diet and cytotoxic chemotherapy substantially raises tumor NADH and synergistically suppresses tumor growth, tripling the survival benefits of chemotherapy alone. Chemotherapy and ketogenic diet also synergize in immune-deficient mice, although long-term growth suppression was only observed in mice with an intact immune system. Conclusions: Ketogenic diet sensitizes murine pancreatic cancer tumors to cytotoxic chemotherapy. Based on these data, we have initiated a randomized clinical trial of chemotherapy with standard versus ketogenic diet for patients with metastatic pancreatic cancer (NCT04631445).


Subject(s)
Diet, Ketogenic , Pancreatic Neoplasms , Animals , Carbohydrates , Diet, Ketogenic/methods , Humans , Mice , NAD , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Pancreatic Neoplasms
3.
J Biochem Mol Toxicol ; 36(2): e22958, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34783111

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is a fatal malignancy and patients with PDAC are mostly diagnosed at advanced stages. Lutein is a natural compound that belongs to the non-vitamin A carotenoids family and has presented antitumor effects on multiple cancer types. However, the function of lutein in PDAC and the mechanisms are not reported. Here, we explored the role of lutein in PDAC progression. Bioinformatic analysis identified that lutein is correlated with PDAC. Lutein suppressed the proliferation, migration, and invasion of PANC-1 cells. The upregulated genes in PDAC patients were identified and the overlap analysis predicted BAG3 as one target of lutein. Lutein repressed BAG3 expression and bioinformatics analysis predicted the interaction between lutein and BAG3. The inhibitory effects of lutein on PANC-1 cell proliferation, migration, and invasion are reversed by overexpression of BAG3. GSEA analysis identified that cholesterol homeostasis as one of the downstream signaling pathways of BAG3. In conclusion, lutein induced an inhibitory effect on the malignant progression of PDAC by targeting BAG3/cholesterol homeostasis. Lutein may be applied as a promising candidate for PDAC therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/metabolism , Apoptosis Regulatory Proteins/metabolism , Cholesterol/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Lutein/pharmacology , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Adenocarcinoma/drug therapy , Cell Line, Tumor , Homeostasis/drug effects , Humans , Pancreatic Neoplasms/diet therapy
4.
Nutrients ; 13(9)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34579079

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and extremely therapy-resistant cancer. It is estimated that up to 80% of PDAC patients present with cachexia, a multifactorial disorder characterized by the involuntary and ongoing wasting of skeletal muscle that affects therapeutic response and survival. During the last decade, there has been an increased interest in exploring dietary interventions to complement the treatment of PDAC and associated cachexia. Ketogenic diets (KDs) have gained attention for their anti-tumor potential. Characterized by a very low carbohydrate, moderate protein, and high fat composition, this diet mimics the metabolic changes that occur in fasting. Numerous studies report that a KD reduces tumor growth and can act as an adjuvant therapy in various cancers, including pancreatic cancer. However, research on the effect and mechanisms of action of KDs on PDAC-associated cachexia is limited. In this narrative review, we summarize the evidence of the impact of KDs in PDAC treatment and cachexia mitigation. Furthermore, we discuss key cellular mechanisms that explain KDs' potential anti-tumor and anti-cachexia effects, focusing primarily on reprogramming of cell metabolism, epigenome, and the gut microbiome. Finally, we provide a perspective on future research needed to advance KDs into clinical use.


Subject(s)
Adenocarcinoma/complications , Adenocarcinoma/diet therapy , Cachexia/etiology , Diet, Ketogenic , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/diet therapy , Humans
5.
Pancreas ; 50(5): 657-666, 2021.
Article in English | MEDLINE | ID: mdl-34106574

ABSTRACT

ABSTRACT: Diet and exercise interventions may help reverse malnutrition and muscle wasting common in pancreatic cancer. We performed a scoping review to identify the knowledge gaps surrounding diet and exercise interventions. We searched PubMed, Scopus, Cumulative Index to Nursing and Allied Health Literature, Embase, ProQuest Theses and Dissertations, and Google Scholar using the umbrella terms of "pancreatic cancer," "diet/nutrition," and "exercise." Included were articles reporting on ambulatory adults with diagnosed pancreatic cancer. Excluded were studies examining prevention and/or risk, animal, or cell lines. Of the 15,708 articles identified, only 62 met the final inclusion criteria. Almost half of the articles were randomized controlled studies (n = 27). Most studies were from the United States (n = 20). The majority examined dietary interventions (n = 41), with 20 assessing the use of omega-3 fatty acids. Exercise interventions were reported in 13 studies, with 8 examining a diet and exercise intervention. Most studies were small and varied greatly in terms of study design, intervention, and outcomes. We identified 7 research gaps that should be addressed in future studies. This scoping review highlights the limited research examining the effect of diet and exercise interventions in ambulatory patients with pancreatic cancer.


Subject(s)
Cachexia/diet therapy , Exercise Therapy , Malnutrition/diet therapy , Muscular Atrophy/diet therapy , Nutrition Therapy , Pancreatic Neoplasms/diet therapy , Body Composition , Cachexia/epidemiology , Cachexia/physiopathology , Diet, Healthy , Dietary Supplements/adverse effects , Humans , Malnutrition/epidemiology , Malnutrition/physiopathology , Muscle Strength , Muscle, Skeletal/physiopathology , Muscular Atrophy/epidemiology , Muscular Atrophy/physiopathology , Nutritional Status , Nutritive Value , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/physiopathology , Treatment Outcome
6.
Nutr. clín. diet. hosp ; 41(2): 61-66, 2021. ilus
Article in Portuguese | IBECS | ID: ibc-225667

ABSTRACT

Introdução: O aumento da incidência de doenças crônicas não transmissíveis e a mortalidade associada a estas causas têm se destacado mundialmente. Entre essas doenças, destaca-se o câncer de pâncreas, que é caracterizado por tendência à evolução com metástase e baixa sobrevida. Relato de caso: As terapias oncológicas podem afetar a qualidade de vida e o estado nutricional dos pacientes e, por essa razão, a utilização de terapias alternativas e complementares, como o uso da própolis, podem auxiliar na melhoria da qualidade do tratamento, através da diminuição na proliferação de células neoplásicas e dos efeitos tóxicos da quimioterapia, devido às características epigenéticas, antitumorais, apoptóticas, antioxidantes e imunomodulatórias. Este relato de caso aborda o acompanhamento clínico e nutricional de um paciente idoso do sexo masculino, portador de câncer pancreático em tratamento quimioterápico, sob aconselhamento nutricional associado à suplementação de extrato hidroalcoólico de própolis verde. Conclusão: Observou-se com este relato de caso, a melhora da qualidade de vida e aumento da taxa de sobrevida do paciente de 12 meses para três anos e meio, além de estabilização da progressão tumoral. (AU)


Introduction: The increase in the incidence of chronic noncommunicable diseases has been highlighted in terms of worldwide mortality rates. Among these diseases, pancreatic cancer stands out, which is characterized by a tendency towards the evolution of metastasis and low survival. Weight loss is associated with increased basal energy expenditure, decreased energy consumption and malabsorption of nutrients. Case report: Oncological therapies can affect to quality of life and nutritional status of individuals, due to the toxic and immunosuppressive effects. For this reason, the use of alternative and complementary therapies, such as the use of propolis, can help to improve the quality of treatment, by decreasing the proliferation of neoplastic cells and the toxic effects of chemotherapy, due to the epigenetic, antitumor, apoptotic characteristics, antioxidants and immunomodulatory. This case report addresses the clinical and nutritional monitoring of an elderly male patient, with pancreatic cancer undergoing chemotherapy, under nutritional advice associated with the supplementation of hydroalcoholic extract of green propolis. Conclusion: There was an improvement in the quality of life and an increase in the patient’s survival rate from 12 months to three years, in addition to stabilization of tumor progression. (AU)


Subject(s)
Humans , Male , Aged , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Dietary Supplements , Propolis , Quality of Life
7.
Sci Rep ; 10(1): 20319, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230218

ABSTRACT

Pancreatic cancer is one of the most lethal and chemo-resistant cancers worldwide. Growing evidence supports the theory that the gut microbiota plays an essential role in modulating the host response to anti-cancer therapy. The present study aimed to explore the effect of probiotics as an adjuvant during chemotherapy for pancreatic cancer. An LSL-KrasG12D/--Pdx-1-Cre mouse model of pancreatic ductal adenocarcinoma (PDAC) was created to study the effects of using four-week multi-strain probiotics (Lactobacillus paracasei GMNL-133 and Lactobacillus reuteri GMNL-89) as an adjuvant therapy for controlling cancer progression. At 12 weeks of age, pancreatitis was induced in the mice by two intraperitoneal injection with caerulein (25 µg/kg 2 days apart). Over the next 4 weeks the mice were treated with intraperitoneal injections of gemcitabine in combination with the oral administration of probiotics. The pancreas was then harvested for analysis. Following caerulein treatment, the pancreases of the LSL-KrasG12D/--Pdx-1-Cre transgenic mice exhibited more extensive pancreatic intraepithelial neoplasia (PanIN) formation. Combined treatment with gemcitabine and probiotics revealed a lower grade of PanIN formation and a decrease in the expression of vimentin and Ki-67. Mice that received gemcitabine in combination with probiotics had lower aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. Notably, the use of high-dose probiotics alone without gemcitabine also had an inhibitory effect on PanIN changes and serum liver enzyme elevation. These findings suggest that probiotics are able to make standard chemotherapy more effective and could help improve the patient's tolerance of chemotherapy.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Pancreatic Ductal/diet therapy , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Lactobacillus , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Probiotics/administration & dosage , Administration, Oral , Animals , Carcinoma, Pancreatic Ductal/chemically induced , Carcinoma, Pancreatic Ductal/microbiology , Ceruletide/adverse effects , Deoxycytidine/administration & dosage , Disease Models, Animal , Female , Gastrointestinal Microbiome/drug effects , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/microbiology , Treatment Outcome , Gemcitabine
8.
Anticancer Res ; 40(2): 873-880, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32014931

ABSTRACT

BACKGROUND/AIM: The acidic tumor microenvironment is associated both with the progression and drug resistance of cancer. We aimed to investigate the effects of alkalization therapy performed concurrently with chemotherapy on the survival of advanced pancreatic cancer patients (study registration: UMIN 000035659). PATIENTS AND METHODS: Twenty-eight patients with metastatic or recurrent pancreatic cancer were assessed in this study. Alkalization therapy consisted of an alkaline diet with supplementary oral sodium bicarbonate (3.0-5.0 g/day). RESULTS: The mean urine pH was significantly higher after the alkalization therapy (6.85±0.74 vs. 6.39±0.92; p<0.05). The median overall survival from the start of alkalization therapy of the patients with high urine pH (>7.0) was significantly longer than those with low urine pH (≤ 7.0) (16.1 vs. 4.7 months; p<0.05). CONCLUSION: An alkalization therapy may be associated with better outcomes in advanced pancreatic cancer patients treated with chemotherapy.


Subject(s)
Neoplasm Recurrence, Local/diet therapy , Neoplasm Recurrence, Local/drug therapy , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/drug therapy , Sodium Bicarbonate/administration & dosage , Aged , Aged, 80 and over , Dietary Supplements , Female , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/urine , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/urine , Retrospective Studies
9.
Article in English | MEDLINE | ID: mdl-32047477

ABSTRACT

Background: Nesidioblastosis and insulinoma are disorders of the endocrine pancreas causing endogenous hyperinsulinemic hypoglycemia. Their coexistence is very unusual and treatment represents a still unresolved dilemma. Case Description: The patient was a 43-year-old Caucasian woman, with a 2-year history of repeated severe hypoglycemic events. The diagnostic work-up was strongly suggestive of insulinoma and the patient was submitted to surgical treatment carried out laparoscopically under robotic assistance. However, surgical exploration and intraoperative ultrasonography failed to detect a pancreatic tumor. Resection was therefore carried out based on the results of selective intra-arterial calcium stimulation test, following a step-up approach, eventually leading to a pancreatoduodenectomy at the splenic artery. The histopathology examination and the immunohistochemical staining were consistent with adult-onset nesidioblastosis. After surgery, the patient continued to experience hypoglycemia with futile response to medical treatments (octreotide, calcium antagonists, diazoxide, and prednisone). Following multidisciplinary evaluation and critical review of a repeat abdominal computed tomography scan, a small nodular lesion was identified in the tail of the pancreas. The nodule was enucleated laparoscopically and the pathological examination revealed an insulinoma. In spite of the insulinoma resection, glycemic values were only partially restored, with residual nocturnal hypoglycemia. Administration of uncooked cornstarch (1.25 g/kg body weight) at bedtime was associated with significant improvement of interstitial glucose levels (p < 0.0001) and reduction of nocturnal hypoglycemia episodes (p = 0.0002). Conclusions: This report describes a rare coexistence of adult-onset nesidioblastosis and insulinoma, suggesting the existence of a wide and continuous spectrum of proliferative ß-cell changes. Moreover, we propose that uncooked cornstarch may offer an additional approach to alleviate the hypoglycemic episodes when surgery is impracticable/unaccepted.


Subject(s)
Insulinoma/complications , Nesidioblastosis/complications , Pancreatic Neoplasms/complications , Adult , Blood Glucose/drug effects , Blood Glucose/metabolism , Circadian Rhythm , Congenital Hyperinsulinism/diagnosis , Congenital Hyperinsulinism/diet therapy , Congenital Hyperinsulinism/etiology , Congenital Hyperinsulinism/surgery , Diagnosis, Differential , Female , Humans , Hypoglycemia/etiology , Hypoglycemia/prevention & control , Insulinoma/diagnosis , Insulinoma/diet therapy , Insulinoma/surgery , Nesidioblastosis/diagnosis , Nesidioblastosis/diet therapy , Nesidioblastosis/surgery , Pancreatectomy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/surgery , Starch/therapeutic use
10.
Medicina (Kaunas) ; 56(2)2020 Jan 24.
Article in English | MEDLINE | ID: mdl-31991566

ABSTRACT

Background and objectives: Immunonutrition is recommended by enhanced recovery after surgery in patients undergoing pancreatoduodenectomy for 5-7 days perioperatively as it may reduce the rate of infectious complications. However, data on effect of immunonutrition on the overall complication rate are contradictory and it is not clear, which groups of patients benefit most. The aims of this study are to evaluate the effects of immunonutrition on the overall complication rate and the rate of severe and/or multiple complications in patients with pancreatic tumours stratified according to final histological diagnosis-patients with pancreatic ductal adenocarcinoma (PDAC) vs. other tumours-and nutritional state, using more sensitive Comprehensive Complication Index. Materials and Methods: Seventy consecutive patients scheduled for pancreatoduodenectomy because of pancreatic tumours were randomised into immunonutrition vs. control groups and stratified according to final histological diagnosis and nutritional status. Surgical outcomes were assessed postoperatively using Clavien-Dindo classification (CDC) and Comprehensive Complication Index (CCI). Results: No significant differences in the overall complication rates in immunonutrition vs. control, patients with malnutrition vs. no malnutrition, PDAC vs. other pancreatic tumours groups were detected. However, significant differences in the rates of severe and/or multiple complications in immunonutrition vs. control groups and in PDAC patients segregated according to immunonutrition were obtained using CCI. Conclusions: Patients with PDAC may experience greater benefits of immunonutrition as compared to patients with benign pancreatic diseases or less aggressive tumours, while nutritional status was not a determining factor for the efficacy of immunonutrition.


Subject(s)
Nutritional Physiological Phenomena , Pancreatic Neoplasms/diet therapy , Pancreaticoduodenectomy/methods , Postoperative Complications/etiology , Aged , Female , Humans , Lithuania/epidemiology , Male , Middle Aged , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/surgery , Postoperative Complications/epidemiology , Prospective Studies
11.
Biomolecules ; 11(1)2020 12 29.
Article in English | MEDLINE | ID: mdl-33383727

ABSTRACT

Dietary patterns are well known risk factors involved in cancer initiation, progression, and in cancer protection. Previous in vitro and in vivo studies underline the link between a diet rich in resistant starch (RS) and slowing of tumor growth and gene expression in pancreatic cancer xenograft mice. The aim of this study was to investigate the impact of a diet rich in resistant starch on miRNAs and miRNAs-target genes expression profile and on biological processes and pathways, that play a critical role in pancreatic tumors of xenografted mice. miRNA expression profiles on tumor tissues displayed 19 miRNAs as dysregulated in mice fed with RS diet as compared to those fed with control diet and differentially expressed miRNA-target genes were predicted by integrating (our data) with a public human pancreatic cancer gene expression dataset (GSE16515). Functional and pathway enrichment analyses unveiled that miRNAs involved in RS diet are critical regulators of genes that control tumor growth and cell migration and metastasis, inflammatory response, and, as expected, synthesis of carbohydrate and glucose metabolism disorder. Mostly, overall survival analysis with clinical data from TCGA (n = 175) displayed that almost four miRNAs (miRNA-375, miRNA-148a-3p, miRNA-125a-5p, and miRNA-200a-3p) upregulated in tumors from mice fed with RS were a predictor of good prognosis for pancreatic cancer patients. These findings contribute to the understanding of the potential mechanisms through which resistant starch may affect cancer progression, suggesting also a possible integrative approach for enhancing the efficacy of existing cancer treatments.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/genetics , Prebiotics , Resistant Starch/therapeutic use , Animals , Cell Line, Tumor , Diet , Female , Humans , Mice, Nude , Pancreatic Neoplasms/diagnosis , Prebiotics/analysis , Prognosis , Resistant Starch/analysis , Survival Analysis , Transcriptome
12.
Nutr Cancer ; 72(2): 202-208, 2020.
Article in English | MEDLINE | ID: mdl-31271302

ABSTRACT

Aim: The purpose of this study was to identify the nutritional and performance status of Pancreatic Cancer (PanCa) patients and to determine the relationship between these parameters and their survival time.Material and Methods: Ninety-six PanCa patients [59.6% F, 61.4% M; mean age: 60.7 (min:28, max:80) years] were followed up for 6-24 months. The Patient Generated Subjective Global Assessment (PG-SGA) and Eastern Comparative Oncology Group (ECOG) scale were performed. Anthropometric measurements [height, weight, mid-upper arm circumference (MUAC), calf circumference (CC) and triceps skin fold thickness (TSF)], hand grip strength (HGS) were recorded. Survival analyses were conducted using Kaplan-Meier curves.Results: Malnutrition was observed in 85.5% (n = 82) and 54.2% of all patients had poor performance status. A positive correlation was observed between malnutrition and ECOG scale of the patients (P < .01). Antropometric measurements for women and men, respectively, were 34.4 ± 3.03-34.6 ± 3.43 cm for CC; 26.9 ± 3.47-26.05 ± 3.37 cm for MUAC; 20.5 ± 6.3-13.02 ± 7.7 mm for TSF; - 31.02 ± 7.64-20.13 ± 6.04 kg for HGS. Survival time of patients with SGA-A and B was 38.0 ± 6.6 months and of those with SGA-C was 12.0 ± 3.1 months (P = .000).Conclusion: Malnutrition negatively affected both performance status and survival time among PanCa patients.


Subject(s)
Anthropometry/methods , Body Mass Index , Body Weight/physiology , Hand Strength , Malnutrition/physiopathology , Pancreatic Neoplasms/mortality , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Nutrition Assessment , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/pathology , Prospective Studies , Survival Rate
13.
Clin Nutr ESPEN ; 34: 116-124, 2019 12.
Article in English | MEDLINE | ID: mdl-31677701

ABSTRACT

BACKGROUNDS AND AIMS: This randomized clinical trial examined efficacy of prolonged elemental diet (ED) therapy after pancreatoduodenectomy (PD) for pancreatic ductal adenocarcinoma (PDAC), which often causes postoperative malnutrition leading to worsened short- and long-term outcomes. METHODS: Thirty-nine patients with PDAC receiving PD was randomly assigned to prolonged ED group (PEDG) and control group (CG). Fat-free ED (Elental®, EA Pharma CO., Ltd., Tokyo, Japan) via tube jejunostomy was initiated on postoperative day 1 and increased to maintain with 600 kcal/day in addition to oral intake. ED was discontinued if sufficient oral intake was achieved in CG but continued during 3 postoperative months in PEDG. Primary outcome was complication necessitating readmission. Secondary outcomes were nutritional parameters, relative dose intensity (RDI) in cases of adjuvant chemotherapy, and survival outcomes. RESULTS: Twenty patients were assigned to CG and 19 to PEDG. Cumulative post-discharge readmission rate was significantly lower in PEDG than in CG (PEDG vs CG; 12.6% vs 43.7% at 12-post-discharge-month; p = 0.018). Total calorie and ED-derived protein intakes were significantly larger in PEDG than in CG up to 3-postoperative-month but thereafter similar among groups. Lymphocyte counts were significantly increased and neutrophil-to-lymphocyte-ratio (NLR) was significantly reduced in PEDG than in CG at 2-, 3-, and 6-postoperative-month. However, other outcome measures did not differ among groups. CONCLUSION: This trial failed to show survival benefit of prolonged ED therapy but demonstrated its favorable effect on increased lymphocyte counts, reduced NLR, and prevention of complications necessitating readmission, those which may lead to survival benefit with some modifications.


Subject(s)
Adenocarcinoma/diet therapy , Food, Formulated , Pancreatic Neoplasms/diet therapy , Pancreaticoduodenectomy , Adult , Aged , Aged, 80 and over , Energy Intake , Female , Humans , Intubation, Gastrointestinal , Japan , Lymphocyte Count , Male , Middle Aged , Nutritional Status , Postoperative Period , Prospective Studies , Treatment Outcome , Pancreatic Neoplasms
14.
Clin Nutr ESPEN ; 33: 148-153, 2019 10.
Article in English | MEDLINE | ID: mdl-31451252

ABSTRACT

BACKGROUND & AIMS: Neoadjuvant chemoradiotherapy (NACRT) for pancreatic cancer (PC) is potentially associated with various toxicities, which can lead to impaired nutritional status. Eicosapentaenoic acid (EPA) can reduce proinflammatory cytokines and positively influence cancer cachexia syndrome. The aim of this study is to clarify the utility of EPA enriched nutrition support during NACRT for PC. METHODS: We randomly assigned 62 patients with PC that received NACRT to either a nutrition intervention (NI) or a normal diet (ND). Patients in the NI group received 2 bottles/day (550 kcal/day) of an EPA-enriched nutrition supplement during NACRT. The primary endpoints were the before-to-after NACRT ratios (post/pre ratios) of skeletal muscle mass and psoas major muscle area (PMA). The secondary endpoints were the post/pre ratios of other nutritional parameters and treatment-related toxicities. RESULTS: Only 14 patients (45.2%) in the NI group consumed more than 50% of the EPA-enriched supplement provided. The post/pre ratio of skeletal muscle mass in the NI group (0.99 ± 0.060) was not significantly different from that of the ND group (0.96 ± 0.079, p = 0.102). However, patients that consumed ≥50% of the EPA-enriched supplement (the good intake group) had significantly higher skeletal muscle mass ratios than patients in the ND group (p = 0.042). The PMA ratio was significantly higher in the NI group (0.96 ± 0.081) than in the ND group (0.89 ± 0.072, p = 0.001). The NI and ND groups were not significantly different in other nutritional parameters or in NACRT-related toxicity. CONCLUSIONS: We found that EPA-enriched intake could potentially improve the nutritional status of patients with PC that received NACRT, but it was difficult for many patients to drink, due to its disagreeable taste. University Hospital Medical Information Network (http://www.umin.ac.jp), registration number UMIN000033589, https://upload.umin.ac.jp/cgi-bin/ctr_e/ctr_view.cgi?recptno=R000038300.


Subject(s)
Chemoradiotherapy/methods , Eicosapentaenoic Acid/therapeutic use , Nutritional Status , Nutritional Support/methods , Pancreatic Neoplasms/diet therapy , Aged , Dietary Supplements , Eicosapentaenoic Acid/analogs & derivatives , Female , Humans , Male , Middle Aged , Muscle, Skeletal , Preoperative Care , Prospective Studies
15.
BMC Cancer ; 19(1): 528, 2019 May 31.
Article in English | MEDLINE | ID: mdl-31151425

ABSTRACT

BACKGROUND: Most advanced elderly cancer patients experience fatigue, anorexia, and declining physical function due to cancer cachexia, for which effective interventions have not been established. We performed a phase I study of a new nonpharmacological multimodal intervention called the nutritional and exercise treatment for advanced cancer (NEXTAC) program and reported the excellent feasibility of and compliance with this program in elderly patients with advanced cancer who were at risk for cancer cachexia. We report here the background, hypothesis, and design of the next-step multicenter, randomized phase II study to evaluate the efficacy of the program, the NEXTAC-TWO study. METHODS: Patients with chemo-naïve advanced non-small cell lung cancer or pancreatic cancer, age ≥ 70 years, performance status ≤2, with adequate organ function and without disability according to the modified Katz index will be eligible. In total, 130 participants will be recruited from 15 Japanese institutions and will be randomized into either the intervention group or a control group. Computer-generated random numbers are allocated to each participant. Stratification factors include performance status (0 to 1 vs. 2), site of primary cancer (lung vs. pancreas), stage (III vs. IV), and type of chemotherapy (cytotoxic vs. others). Interventions and assessment will be performed 4 times every 4 ± 2 weeks from the date of randomization. Interventions will consist of nutritional counseling, nutritional supplements (rich in branched-chain amino acids), and a home-based exercise program. The exercise program will include low-intensity daily muscle training and lifestyle education to promote physical activity. The primary endpoint is disability-free survival. It is defined as the period from the date of randomization to the date of developing disability or death due to any cause. This trial also plans to evaluate the improvements in nutritional status, physical condition, quality of life, activities of daily living, overall survival, and safety as secondary endpoints. Enrollment began in August 2017. The study results will demonstrate the efficacy of multimodal interventions for elderly cancer patients and their application for the maintenance of physical and nutritional conditions in patients with cancer cachexia. This work is supported by a grant-in-aid from the Japan Agency for Medical Research and Development. DISCUSSION: This is the first randomized trial to evaluate the efficacy and safety of a multimodal intervention specific for elderly patients with advanced cancer. TRIAL REGISTRATION: Registered at August 23, 2017. Registry number: UMIN000028801 .


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Pancreatic Neoplasms/therapy , Aged , Aged, 80 and over , Cachexia/epidemiology , Cachexia/physiopathology , Cachexia/prevention & control , Cachexia/therapy , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Protocols , Clinical Trials, Phase II as Topic , Combined Modality Therapy , Exercise Therapy , Humans , Japan , Lung Neoplasms/diet therapy , Lung Neoplasms/pathology , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/pathology , Quality of Life , Randomized Controlled Trials as Topic
16.
Clin Nutr ESPEN ; 30: 94-99, 2019 04.
Article in English | MEDLINE | ID: mdl-30904235

ABSTRACT

BACKGROUND & AIMS: We investigated the predictors of overall survival (OS) among Korean patients with advanced pancreatic cancer (PC) according to their baseline nutritional status. METHODS: We retrospectively reviewed the data of 412 inpatients with PC between January 2007 and February 2015 at the Department of Oncology of the Gangnam Severance Hospital, Korea. Data on demographic and clinical parameters were collected from electronic medical records, and OS was estimated using the Kaplan-Meier method. Stepwise Cox regression analysis was used to determine the factors associated with survival. Patients with a Nutritional Risk Screening (NRS) 2002 score <3 were classified as "no-risk; " those with a score of 3 were classified as "moderate-risk; " and those with a score of ≥4 were classified as "high-risk." RESULTS: Following nutritional screening at baseline, 194 patients (47.1%, mean age 61.8 ± 9.9 years) were classified as the "no risk" group; 81 patients (19.7%, mean age 65.4 ± 10.8 years), as the "moderate risk" group; and 137 patients (33.3%, mean age 67.8 ± 12.0 years), as the "high risk" group. Predictors of survival were NRS 2002 score (hazard ratio [HR] = 1.238; 95% confidence interval [CI] = 1.143-1.341), percentage of lymphocytes (HR = 0.973; 95% CI = 0.962-0.984), C-reactive protein level (HR = 1.003; 95% CI = 1.001-1.006), carcinoembryonic antigen level (HR = 1.000; 95% CI = 1.000-1.000), and carbohydrate antigen 19-9 level (HR = 1.000; 95% CI = 1.000-1.000). Kaplan-Meier survival analysis showed significant differences in the median OS among the NRS 2002 groups: "no risk" group: 12.3 ± 0.4 months (95% CI: 11.47-13.13 months); "moderate risk" group: 6.5 ± 0.9 months (95% CI: 4.78-8.17 months); and "high risk" group: 5.5 ± 0.6 months (95% CI: 4.31-6.69 months). CONCLUSIONS: A good baseline nutritional status was associated with OS among Korean patients with advanced PC. An improvement in the nutritional status of patients with advanced PC through baseline nutritional interventions is therefore necessary to prolong OS.


Subject(s)
Nutritional Status , Pancreatic Neoplasms/mortality , Aged , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/pathology , Prognosis , Republic of Korea , Retrospective Studies , Survival Analysis
17.
Biochem Genet ; 57(4): 477-486, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30649640

ABSTRACT

Pancreatic cancer is the fourth leading cause of death in both males and females, with a 5-year relative survival rate of 8%. The Wnt signaling pathway has a significant role in the pathogenesis of many tumors, including those of pancreatic cancer. Hypermethylation of the Wnt inhibitory Factor-1 (WIF1) gene promoter have been detected in different types of cancer. In contrast, the anticancer effects of long-chain omega-3 PUFA (ALA) have been reported. Regarding its anticancer effects, in this study, we investigated the effects of various concentrations of omega-3 PUFA on expression level and promoter methylation of the WIF1 gene in MIA PaCa-2 cells in 24, 48, and 72 h after treatment. MIA PaCa-2 cells were treated with different concentrations of omega-3 PUFA (25, 50, 100, 250, 500, and 1000 µM). Cell viability assay was carried out followed by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and methylation-specific PCR (MSP). This investigation suggested that dietary consumption of omega-3 PUFAs (250-1000 µM) has a significant effect on the proliferation and WIF1 gene expression of the MIA PaCa-2 cancer cell line but no effect on the promoter methylation of this gene. Changes in promoter methylation were not observed in any of the treatments.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Fatty Acids, Omega-3/administration & dosage , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/genetics , Repressor Proteins/genetics , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
18.
Anticancer Res ; 38(11): 6519-6527, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30396981

ABSTRACT

BACKGROUND/AIM: High-carbohydrate diets are generally provided to post-pancreatectomy cancer patients. Low energy density of this diet may obstruct proper energy intake and recovery. This study aimed to assess the effects of high-fat, high-energy ketogenic diet (KD) in these patients. PATIENTS AND METHODS: After pancreatectomy, 9 patients were provided with general diet (GD) while 10 were served KD. Meal compliance, energy intake rate, meal satisfaction and presence of complications were monitored throughout hospital stay. Data on nutritional status, serum lipids and body composition were collected and compared between groups. RESULTS: Meal compliance, energy intake rate and meal satisfaction score were higher in KD. There were no differences in complications, nutritional status and serum lipids. The decrease in body cell mass (BCM) was greater in GD. CONCLUSION: Post-pancreatectomy cancer patients who consumed KD had a higher energy intake and BCM. These results suggest the potential use of KD as an adjuvant anti-cancer therapy.


Subject(s)
Biliary Tract Neoplasms/diet therapy , Biliary Tract Neoplasms/surgery , Diet, Ketogenic/methods , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/surgery , Adult , Aged , Chemotherapy, Adjuvant , Energy Metabolism , Female , Hospitalization , Humans , Male , Middle Aged , Nutritional Status , Pancreatectomy , Patient Compliance , Patient Satisfaction , Prospective Studies , Treatment Outcome
19.
Nutr Cancer ; 70(7): 1075-1087, 2018 10.
Article in English | MEDLINE | ID: mdl-30273070

ABSTRACT

Pancreatic cancer (PC) patients have poor prognosis and survival rate. Gemcitabine, the drug of choice has a dismal 15% response rate. Earlier, we reported that Garcinol alone and in combination with gemcitabine showed a dose-dependent favorable response on PC cell lines. This study probes the in vivo effects of dietary Garcinol on PC progression in transgenic PC mice (KPC; K-ras and p53 conditional mutant). KPC male mice were divided into: KC- Control diet; KGr-0.05% Garcinol diet; KGm-Gemcitabine injected; KGG - Garcinol diet + Gemcitabine injected groups. Changes in tumor progression, toxicity, or cell morphology were monitored by magnetic resonance imaging, Fore-stomach, and blood smear, respectively. Pancreatic Intraepithelial Neoplasia (mPanIN) grading with hematoxylin and eosin (H&E) staining was conducted on pancreas and validated by immunohistochemistry. The KGr group showed improved survival, no observable toxicity with marked reduction in papilloma formation in the fore-stomach, and a higher ratio of NK and NKT cells compared to Non-NK lymphocytes. Additionally, the KGr, KGm, and KGG groups showed reduction in tumor volumes and reduced number of advanced mouse PanIN3. Dietary Garcinol alone and in combination with gemcitabine retarded the progression of PC in transgenic PC mice, arresting the cancer in the earlier stages, improving prognosis and survival.


Subject(s)
Pancreatic Neoplasms/diet therapy , Terpenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dietary Supplements , Genes, p53 , Genes, ras , Humans , Magnetic Resonance Imaging , Male , Mice, Transgenic , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , S100 Calcium Binding Protein beta Subunit/immunology , Smad4 Protein/immunology , Survival Rate , Terpenes/adverse effects , Gemcitabine
20.
In Vivo ; 32(4): 759-763, 2018.
Article in English | MEDLINE | ID: mdl-29936456

ABSTRACT

BACKGROUND/AIM: From the standpoint of cancer therapy, it is valuable to enhance the anticancer effects of chemotherapy. Our previous reports revealed that up-regulation of heat-shock protein 27 (HSP27) has been linked to gemcitabine resistance of pancreatic cancer cells. Enzyme-treated asparagus extract (ETAS) is an extract that is produced from asparagus. The purpose of this study was to investigate the effect of ETAS on the expression of HSP27 and other HSPs in the gemcitabine-resistant pancreatic cancer cell line KLM1-R. MATERIALS AND METHODS: KLM1-R cells were treated with ETAS, and expression levels of HSPs, including HSP27, were investigated by western blotting. RESULTS: ETAS down-regulated HSP27 and pHSP27 (serine 78) in KLM1-R cells, but, HSP70 and GRP78 levels were not altered. CONCLUSION: This study suggests the potential therapeutic benefit of ETAS in enhancing anticancer effects by its combination with gemcitabine for patients with pancreatic cancer.


Subject(s)
Asparagus Plant/chemistry , Drug Resistance, Neoplasm/drug effects , HSP27 Heat-Shock Proteins/genetics , Pancreatic Neoplasms/diet therapy , Plant Extracts/pharmacology , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/therapeutic use , Cell Line, Tumor , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , HSP72 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Humans , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Plant Extracts/chemistry , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...