Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cancers (Basel) ; 15(16)2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37627143

ABSTRACT

An increased risk of non-pathological fractures in patients with prostate cancer and bone metastases has been associated with combination treatment with radium-223, abiraterone, and prednisone/prednisolone in the absence of bone-protecting agents. Here, we investigated possible mechanisms leading to this outcome using an intratibial LNCaP model mimicking prostate cancer bone metastases. Male NOD.scid mice were inoculated intratibially with LNCaP prostate cancer cells and treated with vehicle, radium-223, abiraterone, prednisone, zoledronic acid, or their combinations for 28 days. Serum TRACP 5b and PSA levels were measured. Bone structure, quality, and formation rate of non-tumor-bearing and tumor-bearing tibiae were analyzed by microCT, 3-point bending assay, and dynamic histomorphometry, respectively. Radium-223 incorporation into bone was also measured. Radium-223/abiraterone/prednisone combination treatment induced a transient increase in bone resorption indicated by elevated TRACP 5b levels, which was inhibited by concurrent treatment with zoledronic acid. Furthermore, radium-223/abiraterone/prednisone combination reduced periosteal and trabecular new bone formation and the number of osteoblasts, but bone structure or biomechanical quality were not affected. The abiraterone/prednisone treatment decreased radium-223 incorporation into tumor-bearing bone, possibly explaining the lack of additional antitumor efficacy. In conclusion, radium-223/abiraterone/prednisone combination increased bone resorption, which may have been one of the mechanisms leading to an increased fracture risk in patients with mCRPC.

2.
Int J Mol Sci ; 24(3)2023 Jan 22.
Article in English | MEDLINE | ID: mdl-36768509

ABSTRACT

Radium-223 dichloride and enzalutamide are indicated for metastatic castration-resistant prostate cancer and their combination is currently being investigated in a large phase 3 clinical trial. Here, we evaluated the antitumor efficacy of radium-223, enzalutamide, and their combination in the intratibial LNCaP model mimicking prostate cancer metastasized to bone. In vitro experiments revealed that the combination of radium-223 and enzalutamide inhibited LNCaP cell proliferation and showed synergistic efficacy. The combination of radium-223 and enzalutamide also demonstrated enhanced in vivo antitumor efficacy, as determined by measuring serum PSA levels in the intratibial LNCaP model. A decreasing trend in the total area of tumor-induced abnormal bone was associated with the combination treatment. The serum levels of the bone formation marker PINP and the bone resorption marker CTX-I were lowest in the combination treatment group and markedly decreased compared with vehicle group. Concurrent administration of enzalutamide did not impair radium-223 uptake in tumor-bearing bone or the ability of radium-223 to inhibit tumor-induced abnormal bone formation. In conclusion, combination treatment with radium-223 and enzalutamide demonstrated enhanced antitumor efficacy without compromising the integrity of healthy bone. The results support the ongoing phase 3 trial of this combination.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms, Castration-Resistant , Radium , Male , Humans , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/pathology , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Radium/therapeutic use , Benzamides/therapeutic use , Nitriles/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/radiotherapy , Bone Neoplasms/pathology
3.
Int J Mol Sci ; 22(11)2021 May 25.
Article in English | MEDLINE | ID: mdl-34070363

ABSTRACT

Osteolytic bone disease is a hallmark of multiple myeloma (MM) mediated by MM cell proliferation, increased osteoclast activity, and suppressed osteoblast function. The proteasome inhibitor bortezomib targets MM cells and improves bone health in MM patients. Radium-223 dichloride (radium-223), the first targeted alpha therapy approved, specifically targets bone metastases, where it disrupts the activity of both tumor cells and tumor-supporting bone cells in mouse models of breast and prostate cancer bone metastasis. We hypothesized that radium-223 and bortezomib combination treatment would have additive effects on MM. In vitro experiments revealed that the combination treatment inhibited MM cell proliferation and demonstrated additive efficacy. In the systemic, syngeneic 5TGM1 mouse MM model, both bortezomib and radium-223 decreased the osteolytic lesion area, and their combination was more effective than either monotherapy alone. Bortezomib decreased the number of osteoclasts at the tumor-bone interface, and the combination therapy resulted in almost complete eradication of osteoclasts. Furthermore, the combination therapy improved the incorporation of radium-223 into MM-bearing bone. Importantly, the combination therapy decreased tumor burden and restored body weights in MM mice. These results suggest that the combination of radium-223 with bortezomib could constitute a novel, effective therapy for MM and, in particular, myeloma bone disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Multiple Myeloma , Neoplasms, Experimental , Animals , Bortezomib/pharmacology , Cell Line, Tumor , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Radioisotopes/pharmacology , Radium/pharmacology
4.
Sci Rep ; 7(1): 5294, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28706193

ABSTRACT

Obesity and dyslipidemia are hallmarks of metabolic and cardiovascular diseases. Polydextrose (PDX), a soluble fiber has lipid lowering effects. We hypothesize that PDX reduces triglycerides and cholesterol by influencing gut microbiota, which in turn modulate intestinal gene expression. C57BL/6 male mice were fed a Western diet (WD) ±75 mg PDX twice daily by oral gavage for 14 days. Body weight and food intake were monitored daily. Fasting plasma lipids, caecal microbiota and gene expression in intestine and liver were measured after 14 days of feeding. PDX supplementation to WD significantly reduced food intake (p < 0.001), fasting plasma triglyceride (p < 0.001) and total cholesterol (p < 0.05). Microbiome analysis revealed that the relative abundance of Allobaculum, Bifidobacterium and Coriobacteriaceae taxa associated with lean phenotype, increased in WD + PDX mice. Gene expression analysis with linear mixed-effects model showed consistent downregulation of Dgat1, Cd36, Fiaf and upregulation of Fxr in duodenum, jejunum, ileum and colon in WD + PDX mice. Spearman correlations indicated that genera enriched in WD + PDX mice inversely correlated with fasting lipids and downregulated genes Dgat1, Cd36 and Fiaf while positively with upregulated gene Fxr. These results suggest that PDX in mice fed WD promoted systemic changes via regulation of the gut microbiota and gene expression in intestinal tract.


Subject(s)
Cholesterol/blood , Diet, Western , Gastrointestinal Microbiome , Glucans/pharmacology , Intestines/physiology , Liver/metabolism , Triglycerides/blood , Animals , Eating , Fasting , Gene Expression Profiling , Intestines/drug effects , Intestines/microbiology , Liver/drug effects , Liver/microbiology , Male , Mice , Mice, Inbred C57BL
5.
Clin Cancer Res ; 23(15): 4335-4346, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28364014

ABSTRACT

Purpose: Radium-223 dichloride (radium-223, Xofigo), a targeted alpha therapy, is currently used for the treatment of patients with castration-resistant prostate cancer (CRPC) with bone metastases. This study examines the mode-of-action and antitumor efficacy of radium-223 in two prostate cancer xenograft models.Experimental Design: Mice bearing intratibial LNCaP or LuCaP 58 tumors were randomized into groups (n = 12-17) based on lesion grade and/or serum PSA level and administered radium-223 (300 kBq/kg) or vehicle, twice at 4-week intervals. X-rays and serum samples were obtained biweekly. Soft tissue tumors were observed macroscopically at sacrifice. Tibiae were analyzed by gamma counter, micro-CT, autoradiography and histology.Results: Radium-223 inhibited tumor-induced osteoblastic bone growth and protected normal bone architecture, leading to reduced bone volume in LNCaP and abiraterone-resistant LuCaP 58 models. Furthermore, radium-223 resulted in lower PSA values and reduced total tissue and tumor areas, indicating that treatment constrains prostate cancer growth in bone. In addition, radium-223 suppressed abnormal bone metabolic activity as evidenced by decreased number of osteoblasts and osteoclasts and reduced level of the bone formation marker PINP. Mode-of-action studies revealed that radium-223 was deposited in the intratumoral bone matrix. DNA double-strand breaks were induced in cancer cells within 24 hours after radium-223 treatment, and PSA levels were significantly lower 72 hours after treatment, providing further evidence of the antitumor effects.Conclusions: Taken together, radium-223 therapy exhibits a dual targeting mode-of-action that induces tumor cell death and suppresses tumor-induced pathologic bone formation in tumor microenvironment of osseous CRPC growth in mice. Clin Cancer Res; 23(15); 4335-46. ©2017 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Bone Neoplasms/radiotherapy , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radium/administration & dosage , Animals , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Bone and Bones/pathology , Bone and Bones/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Disease Models, Animal , Humans , Male , Mice , Osteoclasts/radiation effects , Prostatic Neoplasms, Castration-Resistant/pathology , Radioisotopes/administration & dosage , Tumor Microenvironment/radiation effects
6.
Appetite ; 110: 15-24, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27916475

ABSTRACT

Polydextrose (PDX) reduces subsequent energy intake (EI) when administered at midmorning in single-blind trials of primarily normal-weight men. However, it is unclear if this effect also occurs when PDX is given at breakfast time. Furthermore, for ecological validity, it is desirable to study a female population, including those at risk for obesity. We studied the effects of PDX, served as part of a breakfast or midmorning preload, on subsequent EI and other appetite-related parameters in healthy normal-weight and overweight females. Per earlier studies, the primary outcome was defined as the difference in subsequent EI when PDX was consumed at midmorning versus placebo. Thirty-two volunteers were enrolled in this acute, double-blind, placebo-controlled, randomized, and crossover trial to examine the effects of 12.5 g of PDX, administered as part of a breakfast or midmorning preload, on subsequent EI, subjective feelings of appetite, well-being, and mood. Gastric emptying rates and the blood concentrations of glucose, insulin, cholecystokinin, ghrelin, glucagon-like peptide 1 (GLP-1), and peptide tyrosine-tyrosine were measured in the group that received PDX as part of their breakfast. There were no differences in EI between volunteers who were fed PDX and placebo. PDX intake with breakfast tended to elevate blood glucose (P = 0.06) during the postabsorptive phase, significantly lowered insulin by 15.7% (P = 0.04), and increased GLP-1 by 39.9% (P = 0.02); no other effects on blood parameters or gastric emptying rates were observed. PDX intake at midmorning reduced hunger by 31.4% during the satiation period (P = 0.02); all other subjective feelings of appetite were unaffected. Volunteers had a uniform mood profile during the study. PDX was well tolerated, causing one mild adverse event throughout the trial.


Subject(s)
Appetite/drug effects , Eating/drug effects , Food Additives/administration & dosage , Glucans/administration & dosage , Overweight/drug therapy , Adult , Blood Glucose/analysis , Breakfast/drug effects , Breakfast/psychology , Cholecystokinin/blood , Cross-Over Studies , Dipeptides/blood , Double-Blind Method , Energy Intake/drug effects , Female , Gastric Emptying , Ghrelin/blood , Glucagon-Like Peptide 1/blood , Humans , Hunger/drug effects , Overweight/psychology , Postprandial Period , Satiation/drug effects , Young Adult
7.
Food Chem Toxicol ; 92: 117-28, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27016492

ABSTRACT

AB-LIFE(®) is a probiotic product consisting of equal parts of three strains of Lactobacillus plantarum (CECT 7527, 7528, and 7529) blended with inert excipients. Whole genome sequencing was performed on each of the three strains. Antibiotic resistance was evaluated by genomic mining for resistance genes, and assessment for transferability. No risk of transfer potential was identified for any antibiotic resistance genes in the three strains. AB-LIFE(®) was evaluated for potential subchronic oral toxicity in rats, with dosages of 300 and 1000 mg/kg BW/day (equivalent to 5.55 × 10(10) and 1.85 × 10(11) CFU/kg BW/day). Survival of the three test strains through the gastrointestinal tract was supported by fecal analysis. No adverse effects were identified with respect to in-life parameters, clinical or anatomic pathology, translocation, or fecal chemical analyses. The no-observed-adverse-effect level (NOAEL) for AB-LIFE(®) in male and female rats was 1000 mg/kg BW/day (1.85 × 10(11) CFU of AB-LIFE(®)/kg BW/day), the highest dose level evaluated. These results, in conjunction with a previous acute toxicity study in rats, support the conclusion that AB-LIFE(®) is safe for human consumption.


Subject(s)
Drug Resistance, Microbial/genetics , Feces/microbiology , Gastrointestinal Tract/drug effects , Gene Expression Regulation, Bacterial/drug effects , Lactobacillus plantarum/physiology , Probiotics/toxicity , Toxicity Tests, Subchronic/methods , Administration, Oral , Animals , Feces/chemistry , Female , Genes, Bacterial/genetics , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Male , No-Observed-Adverse-Effect Level , Rats , Safety
8.
Nutrients ; 8(1)2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26784221

ABSTRACT

INTRODUCTION: Subjective feelings of appetite are measured using visual analogue scales (VAS) in controlled trials. However, the methods used to analyze VAS during the Satiation (pre- to post-meal) and Satiety (post-meal to subsequent meal) periods vary broadly, making it difficult to compare results amongst independent studies testing the same product. This review proposes a methodology to analyze VAS during both the Satiation and Satiety periods, allowing us to compare results in a meta-analysis. METHODS: A methodology to express VAS results as incremental areas under the curve (iAUC) for both the Satiation and Satiety periods is proposed using polydextrose as a case study. Further, a systematic review and meta-analysis on subjective feelings of appetite was conducted following the PRISMA methodology. Meta-analyses were expressed as Standardized Mean Difference (SMD). RESULTS: Seven studies were included in the meta-analysis. There were important differences in the methods used to analyze appetite ratings amongst these studies. The separate subjective feelings of appetite reported were Hunger, Satisfaction, Fullness, Prospective Food Consumption, and the Desire to Eat. The method proposed here allowed the results of the different studies to be homogenized. The meta-analysis showed that Desire to Eat during the Satiation period favors polydextrose for the reduction of this subjective feeling of appetite (SMD = 0.24, I² < 0.01, p = 0.018); this effect was also significant in the sub-analysis by sex for the male population (SMD = 0.35, I² < 0.01, p = 0.015). There were no other significant results. CONCLUSION: It is possible to compare VAS results from separate studies. The assessment of iAUC for both the Satiation and Satiety periods generates results of homogeneous magnitudes. This case study demonstrates, for the first time, that polydextrose reduces the Desire to Eat during the Satiation period. This may explain, at least in part, the observed effects of polydextrose on the reduction of levels of energy intake at subsequent meals.


Subject(s)
Appetite/drug effects , Food Additives/pharmacology , Glucans/pharmacology , Postprandial Period/drug effects , Satiation/drug effects , Adult , Appetite/physiology , Area Under Curve , Emotions/drug effects , Female , Humans , Hunger/drug effects , Male , Middle Aged , Randomized Controlled Trials as Topic , Research Design , Satiation/physiology , Visual Analog Scale
9.
Nutr J ; 14: 23, 2015 Mar 08.
Article in English | MEDLINE | ID: mdl-25889643

ABSTRACT

BACKGROUND: Three independent trials were conducted to evaluate postprandial triglyceride (TG) responses in subjects with different lipid metabolism. The effect of polydextrose (PDX), a soluble non-digestible carbohydrate, on postprandial response was also studied using practically relevant, high fat meal interventions. METHODS: A total of 19 normolipidemic (average BMI 24.1 kg/m(2)), 21 overweight/hyperlipidemic (average BMI 29.6 kg/m(2)) and 18 obese/non-diabetic subjects (average BMI 33.6 kg/m(2)) were included in the study. On two separate occasions all subjects ate two high-fat meals (4293 kJ, 36% from fat), one with PDX (either 12.5 g or 15 g) and one without PDX during placebo-controlled, double-blind, crossover and randomized trials. To obtain the triglyceride measurements venous blood samples were taken before the consumption of the test meal and five times afterwards, up to 6 h post-test meal. The triglyceride responses were modeled using a mixed-effects linear model. RESULTS: The key variables that explain the variation of the postprandial triglyceride response in the different subject groups were: baseline triglyceride concentration, time point, and PDX vs. placebo treatment (p < 0.05). The maximum postprandial TG concentration was more pronounced in hyperlipidemic group compared to normolipidemic (p < 0.001) or obese groups (p < 0.01). The modeled TG response analysis showed that irrespective of the study population PDX supplementation was one of the factors significantly reducing triglyceride response compared to the placebo treatment (p < 0.05). CONCLUSIONS: Subjects with elevated fasting triglyceride levels display exaggerated and prolonged postprandial triglyceride responses. PDX, a soluble non-digestible carbohydrate, may offer a dietary concept for reducing the postprandial triglyceride response after the consumption of a meal containing a high concentration of fat.


Subject(s)
Diet, High-Fat/adverse effects , Glucans/pharmacology , Hyperlipidemias/diet therapy , Lipid Metabolism/drug effects , Obesity/diet therapy , Triglycerides/blood , Adult , Body Mass Index , Cross-Over Studies , Double-Blind Method , Female , Food Additives/administration & dosage , Food Additives/pharmacology , Glucans/administration & dosage , Humans , Hyperlipidemias/blood , Hyperlipidemias/metabolism , Male , Middle Aged , Obesity/blood , Obesity/metabolism , Postprandial Period/drug effects , Treatment Outcome , Young Adult
10.
Nutr J ; 14: 2, 2015 Jan 03.
Article in English | MEDLINE | ID: mdl-25555562

ABSTRACT

BACKGROUND: Dietary fibers are associated with enhanced satiety. However, the mechanism of different dietary fibers contributing to satiety-related gastrointestinal (GI) peptide release, especially in an obese population, is still poorly understood. Polydextrose (PDX), a water-soluble glucose polymer, has demonstrated its ability to reduce energy intake at a subsequent meal, but its mechanism of action requires further research. Also, there is limited evidence on its capacity to regulate subjective feelings of appetite. This study examines the effects of PDX on postprandial secretion of satiety-related GI peptides, short chain fatty acids (SCFAs), lactic acid, and subjective appetite ratings in obese participants. METHODS: 18 non-diabetic, obese participants (42.0 y, 33.6 kg/m2) consumed a high-fat meal (4293 kJ, 36% from fat) with or without PDX (15 g) in an acute, multicenter, randomized, double-blind, placebo-controlled and crossover trial. Postprandial plasma concentrations of satiety-related peptides, namely ghrelin, cholecystokinin (CCK), glucagon-like peptide 1 (GLP-1), and peptide YY (PYY), as well as SCFAs and lactic acid were assessed. GI peptide, SCFA and lactate concentrations were then modeled using a linear mixed-effects model.The subjective feelings of hunger, satisfaction, and desire to eat were evaluated using visual analogue scales (VAS), which were analyzed as incremental areas under the curve (iAUC) during the satiation and satiety periods. RESULTS: We found that PDX supplementation increased plasma GLP-1 levels more than the placebo treatment (P = 0.02). In the whole group, GLP-1 concentrations found in participants older than 40 years old were significantly lower (P = 0.01) as compared to those aged 40 years or less. There were no statistically significant differences in postprandial ghrelin, CCK, or PYY responses. The lactic acid concentrations were significantly (P = 0.01) decreased in the PDX group, while no significant changes in SCFAs were found. PDX reduced iAUC for hunger by 40% (P = 0.03) and marginally increased satisfaction by 22.5% (P = 0.08) during the post-meal satiety period. CONCLUSION: Polydextrose increased the postprandial secretion of the satiety hormone GLP-1 and reduced hunger after a high-fat meal. PDX also reduced the elevated postprandial lactic acid levels in plasma. Therefore, PDX may offer an additional means to regulate inter-meal satiety and improve postprandial metabolism in obese participants.


Subject(s)
Glucagon-Like Peptide 1/blood , Glucans/pharmacology , Obesity/physiopathology , Postprandial Period , Satiation/drug effects , Adult , Age Factors , Body Mass Index , Cholecystokinin/blood , Cross-Over Studies , Dietary Fats/administration & dosage , Dietary Fiber/administration & dosage , Double-Blind Method , Fatty Acids, Volatile/blood , Female , Ghrelin/blood , Humans , Lactic Acid/blood , Male , Middle Aged , Peptide YY/blood , Placebos
11.
Appetite ; 87: 30-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25510531

ABSTRACT

INTRODUCTION: Dietary fibers help to control energy intake and reduce the risk of developing obesity. Recent studies show that the consumption of polydextrose reduces energy intake at a subsequent meal. In this systematic review and meta-analysis we examine the subsequent effects of polydextrose on different levels of energy intake (EI). METHOD: The review followed the PRISMA methodology. Meta-analyses were expressed as Standardized Mean Difference (SMD). A linear regression approach was used to model the relationship between the polydextrose dose and the different levels of EI expressed as a relative change (%). RESULTS: All the studies included in this review administered polydextrose as part of a mid-morning snack. Six studies were included in the analysis of EI at an ad libitum lunch; and three were included in the analysis of EI during the rest of the day, as well as total daily EI. The meta-analysis showed that the consumption of polydextrose is associated with a reduction in EI at lunch time (SMD = 0.35; P <0.01; I(2) = 0). The dose of polydextrose consumed correlated significantly with this reduction in EI, EILunch (%) = -0.67 Polydextrose (g/day) (R(2) = 0.80; P <0.01). The meta-analysis of EI during the rest of the day and daily EI did not show any difference. Nevertheless, the regression equation indicates that there is a dose-dependent effect on the reduction of daily EI, EIDaily (%) = -0.35 × Polydextrose (g/day) (R(2) = 0.68; P <0.05). Sex-specific results are consistent with results for the whole group. CONCLUSION: The studies included in this meta-analysis support the notion that the consumption of polydextrose reduces voluntary energy intake at a subsequent meal. Furthermore, this reduction in energy intake occurs in a dose-dependent manner.


Subject(s)
Appetite/drug effects , Diet , Dietary Fiber/pharmacology , Eating/drug effects , Energy Intake/drug effects , Glucans/pharmacology , Meals , Humans
12.
Br J Nutr ; 111(11): 1945-56, 2014 Jun 14.
Article in English | MEDLINE | ID: mdl-24661576

ABSTRACT

Prebiotics, probiotics and synbiotics are dietary ingredients with the potential to influence health and mucosal and systemic immune function by altering the composition of the gut microbiota. In the present study, a candidate prebiotic (xylo-oligosaccharide, XOS, 8 g/d), probiotic (Bifidobacterium animalis subsp. lactis Bi-07, 109 colony-forming units (CFU)/d) or synbiotic (8 g XOS+109 CFU Bi-07/d) was given to healthy adults (25-65 years) for 21 d. The aim was to identify the effect of the supplements on bowel habits, self-reported mood, composition of the gut microbiota, blood lipid concentrations and immune function. XOS supplementation increased mean bowel movements per d (P= 0·009), but did not alter the symptoms of bloating, abdominal pain or flatulence or the incidence of any reported adverse events compared with maltodextrin supplementation. XOS supplementation significantly increased participant-reported vitality (P= 0·003) and happiness (P= 0·034). Lowest reported use of analgesics was observed during the XOS+Bi-07 supplementation period (P= 0·004). XOS supplementation significantly increased faecal bifidobacterial counts (P= 0·008) and fasting plasma HDL concentrations (P= 0·005). Bi-07 supplementation significantly increased faecal B. lactis content (P= 0·007), lowered lipopolysaccharide-stimulated IL-4 secretion in whole-blood cultures (P= 0·035) and salivary IgA content (P= 0·040) and increased IL-6 secretion (P= 0·009). XOS supplementation resulted in lower expression of CD16/56 on natural killer T cells (P= 0·027) and lower IL-10 secretion (P= 0·049), while XOS and Bi-07 supplementation reduced the expression of CD19 on B cells (XOS × Bi-07, P= 0·009). The present study demonstrates that XOS induce bifidogenesis, improve aspects of the plasma lipid profile and modulate the markers of immune function in healthy adults. The provision of XOS+Bi-07 as a synbiotic may confer further benefits due to the discrete effects of Bi-07 on the gut microbiota and markers of immune function.


Subject(s)
Bifidobacterium/metabolism , Glucuronates/administration & dosage , Immune System , Oligosaccharides/administration & dosage , Synbiotics/administration & dosage , Adult , Biomarkers/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Colony Count, Microbial , Cross-Over Studies , Defecation , Double-Blind Method , Feces/microbiology , Female , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Glucuronates/chemistry , Healthy Volunteers , Humans , Immunoglobulin A/metabolism , Interleukin-10/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Oligosaccharides/chemistry , Prebiotics/administration & dosage , Probiotics/administration & dosage , Surveys and Questionnaires , Triglycerides/blood
13.
J Natl Cancer Inst ; 105(12): 908-16, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23682134

ABSTRACT

BACKGROUND: Bone metastases are associated with increased morbidity and poor prognosis in breast cancer patients. Radium-223 dichloride is a calcium mimetic that localizes to bone, providing targeted therapy for skeletal metastasis. METHODS: We investigated the mode of action of radium-223 dichloride using breast cancer cell, osteoclast, and osteoblast cultures as well as a mouse model of breast cancer bone metastasis. A single dose of radium-223 dichloride was used in three different settings mimicking the prevention or treatment of bone metastasis. Disease progression was monitored using fluorescence and radiographic imaging and histological analyses. The effect of radium-223 dichloride alone and in combination with doxorubicin or zoledronic acid on survival of mice was analyzed by Kaplan-Meier methods. All statistical tests used were two-sided. RESULTS: Radium-223 dichloride incorporated into bone matrix and inhibited proliferation of breast cancer cells and differentiation of osteoblasts and osteoclasts (all P values < .001) in vitro. In an established bone metastasis setting, radium-223 dichloride prevented tumor-induced cachexia (0/14 vs 7/14 control mice) and decreased osteolysis by 56% and tumor growth by 43% (all P values < .05). Radium-223 dichloride induced double-strand DNA breaks in cancer cells in vivo. Finally, radium-223 dichloride extended survival as a monotherapy (29.2 days, 95% confidence interval [CI] = 26.6 to 31.8 days, P = .039) and in combination with zoledronic acid (31.4 days, 95% CI = 28.8 to 34.0 days, P = .004) or doxorubicin (31.5 days, 95% CI = 29.5 to 33.5 days, P < .001) compared to the vehicle group (24.9 days, 95% CI = 23.4 to 26.4 days). Similar but even more pronounced effects were observed when radium-223 dichloride was administered in a preventive or micrometastatic setting. CONCLUSIONS: Our findings strongly support the development of radium-223 dichloride for the treatment of breast cancer patients with or at high risk of developing bone metastases.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cachexia/prevention & control , DNA, Neoplasm/drug effects , Radium/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Density Conservation Agents/administration & dosage , Bone Neoplasms/complications , Bone Neoplasms/metabolism , Bone Neoplasms/prevention & control , Cachexia/diagnosis , Cachexia/etiology , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded/drug effects , Diphosphonates/administration & dosage , Disease Models, Animal , Doxorubicin/administration & dosage , Female , Imidazoles/administration & dosage , Kaplan-Meier Estimate , Mice , Osteoblasts/drug effects , Osteoclasts/drug effects , Radioisotopes/pharmacology , Radioisotopes/therapeutic use , Radium/therapeutic use , Stem Cells/drug effects , Tumor Cells, Cultured , Zoledronic Acid
14.
J Nutr Sci ; 2: e44, 2013.
Article in English | MEDLINE | ID: mdl-25191600

ABSTRACT

Elderly adults have alterations in their gut microbiota and immune functions that are associated with higher susceptibility to infections and metabolic disorders. Probiotics and prebiotics, and their synbiotic combinations are food supplements that have been shown to improve both gut and immune function. The objective of this randomised, double-blind, placebo-controlled, cross-over human clinical trial was to study immune function and the gut microbiota in healthy elderly adults. Volunteers (n 37) consumed prebiotic galacto-oligosaccharides (GOS; 8 g/d), probiotic Bifidobacterium lactis Bi-07 (Bi-07; 10(9) colony-forming units/d), their combination (Bi-07 + GOS) and maltodextrin control (8 g/d) in four 3-week periods separated by 4-week wash-out periods. Immune function was analysed by determining the phagocytic and oxidative burst activity of monocytes and granulocytes, whole-blood response to lipopolysaccharide, plasma chemokine concentrations and salivary IgA levels. Gut microbiota composition and faecal SCFA content were determined using 16S ribosomal RNA fluorescence in situ hybridisation and HPLC, respectively. Primary statistical analyses indicated the presence of carry-over effects and thus measurements from only the first supplementation period were considered valid. Subsequent statistical analysis showed that consumption of Bi-07 improved the phagocytic activity of monocytes (P < 0·001) and granulocytes (P = 0·02). Other parameters were unchanged. We have for the first time shown that the probiotic Bi-07 may provide health benefits to elderly individuals by improving the phagocytic activity of monocytes and granulocytes. The present results also suggest that in the elderly, the effects of some probiotics and prebiotics may last longer than in adults.

15.
Clin Cancer Res ; 18(16): 4385-96, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22745104

ABSTRACT

PURPOSE: Preclinical tumor growth experiments often result in heterogeneous datasets that include growing, regressing, or stable growth profiles in the treatment and control groups. Such confounding intertumor variability may mask the true treatment effects especially when less aggressive treatment alternatives are being evaluated. EXPERIMENTAL DESIGN: We developed a statistical modeling approach in which the growing and poorly growing tumor categories were automatically detected by means of an expectation-maximization algorithm coupled within a mixed-effects modeling framework. The framework is implemented and distributed as an R package, which enables model estimation and statistical inference, as well as statistical power and precision analyses. RESULTS: When applied to four tumor growth experiments, the modeling framework was shown to (i) improve the detection of subtle treatment effects in the presence of high within-group tumor variability; (ii) reveal hidden tumor subgroups associated with established or novel biomarkers, such as ERß expression in a MCF-7 breast cancer model, which remained undetected with standard statistical analysis; (iii) provide guidance on the selection of sufficient sample sizes and most informative treatment periods; and (iv) offer flexibility to various cancer models, experimental designs, and treatment options. Model-based testing of treatment effect on the tumor growth rate (or slope) was shown as particularly informative in the preclinical assessment of treatment alternatives based on dietary interventions. CONCLUSIONS: In general, the modeling framework enables identification of such biologically significant differences in tumor growth profiles that would have gone undetected or had required considerably higher number of animals when using traditional statistical methods.


Subject(s)
Models, Statistical , Algorithms , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Computer Simulation , Disease Models, Animal , Humans , MCF-7 Cells , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
J Neurosci Methods ; 182(1): 110-22, 2009 Aug 30.
Article in English | MEDLINE | ID: mdl-19523488

ABSTRACT

Segmentation of Magnetic Resonance multi-layer images of premature infant brain has additional challenges in comparison to normal adult brain segmentation. Images of premature infants contain lower signal to noise ratio due to shorter scanning times. Further, anatomic structure include still greater variations which can impair the accuracy of standard brain models. A fully automatic brain segmentation method for T1-weighted images is proposed in present paper. The method uses watershed segmentation with Gaussian mixture model clustering for segmenting cerebrospinal fluid from brain matter and other head tissues. The effect of the myelination process is considered by utilizing information from T2-weighted images. The performance of the new method is compared voxel-by-voxel to the corresponding expert segmentation. The proposed method is found to produce more uniform results in comparison to three accustomary segmentation methods originally developed for adults. This is the case in particular when anatomic forms are still under development and differ in their form from those of adults.


Subject(s)
Algorithms , Brain/anatomy & histology , Image Interpretation, Computer-Assisted/methods , Infant, Premature , Magnetic Resonance Imaging/methods , Nerve Fibers, Myelinated/ultrastructure , Pattern Recognition, Automated/methods , Computer Simulation , Humans , Image Enhancement/methods , Infant, Newborn , Models, Biological , Models, Statistical , Normal Distribution , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...