Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Expert Opin Investig Drugs ; 30(9): 923-930, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34427483

ABSTRACT

INTRODUCTION: Osteoarthritis (OA) is a serious and incurable disease leading the disability. Surgical treatment is the last but not necessarily the best approach for patients with high risks and costs. However, there are no disease-modifying OA drugs (DMOADs) developed for the disease so far, leaving a huge unmet need for drug treatments. Sprifermin is a recombinant human fibroblast growth factor 18 (rhFGF18) and has been confirmed to have anabolic effects on articular cartilage, which makes it a promising DMOAD. AREAS COVERED: The content of this review includes overview of the market, discovery and development, molecular mechanism, preclinical studies, clinical efficacy, safety, and tolerability of sprifermin. It examines the potential of sprifermin as a disease modifying drug for the treatment of knee OA. EXPERT OPINION: Sprifermin could be one of the most promising DMOADs, especially for cartilage phenotype. Current studies show good tolerability and no safety concerns. Well-designed phase 3 clinical trials are required to examine its effects on symptoms and cartilage loss in knee OA.


Subject(s)
Antirheumatic Agents/administration & dosage , Fibroblast Growth Factors/administration & dosage , Osteoarthritis, Knee/drug therapy , Animals , Antirheumatic Agents/adverse effects , Antirheumatic Agents/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Drug Development , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/pharmacology , Humans , Osteoarthritis, Knee/pathology
2.
Clin Transl Sci ; 14(1): 11-19, 2021 01.
Article in English | MEDLINE | ID: mdl-32583961

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent, dynamic disease that occurs across the age spectrum and can lead to cirrhosis and hepatocellular carcinoma. There are currently no US Food and Drug Administration (FDA) approved treatments for NAFLD; however, this is a field of active research. This review summarizes emerging pharmacotherapies for the treatment of adult and pediatric NAFLD. Investigated pharmacotherapies predominantly target bile acid signaling, insulin resistance, and lipid handling within the liver. Three drugs have gone on to phase III trials for which results are available. Of those, obeticholic acid is the single agent that demonstrates promise according to the interim analyses of the REGENERATE trial. Obeticholic acid showed reduction of fibrosis in adults with nonalcoholic steatohepatitis (NASH) taking 25 mg daily for 18 months (n = 931, reduction in fibrosis in 25% vs. 12% placebo, P < 0.01). Ongoing phase III trials include REGENERATE and MAESTRO-NASH, which investigates thyroid hormone receptor-ß agonist MGL-3196. Outcomes of promising phase II trials in adults with NASH are also available and those have investigated agents, including the fibroblast growth factor (FGF)19 analogue NGM282, the GLP1 agonist liraglutide, the FGF21 analogue Pegbelfermin, the sodium glucose co-transporter 2 inhibitor Empagliflozin, the ketohexokinase inhibitor PF-06835919, the acetyl-coenzyme A carboxylase inhibitor GS-0976, and the chemokine receptor antagonist Cenicriviroc. Completed and ongoing clinical trials emphasize the need for a more nuanced understanding of the phenotypes of subgroups within NAFLD that may respond to an individualized approach to pharmacotherapy.


Subject(s)
Liver Cirrhosis/drug therapy , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Adult , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/adverse effects , Chenodeoxycholic Acid/administration & dosage , Chenodeoxycholic Acid/adverse effects , Chenodeoxycholic Acid/analogs & derivatives , Child , Clinical Trials, Phase III as Topic , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/analogs & derivatives , Glucosides/administration & dosage , Glucosides/adverse effects , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Isobutyrates/administration & dosage , Isobutyrates/adverse effects , Liraglutide/administration & dosage , Liraglutide/adverse effects , Liver/pathology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/pathology , Oxazoles/administration & dosage , Oxazoles/adverse effects , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Pyridazines/administration & dosage , Pyridazines/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Severity of Illness Index , Sulfoxides/administration & dosage , Sulfoxides/adverse effects , Treatment Outcome , Uracil/administration & dosage , Uracil/adverse effects , Uracil/analogs & derivatives
3.
Expert Opin Investig Drugs ; 29(2): 125-133, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31899984

ABSTRACT

Introduction: Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide and is strongly associated with obesity and insulin resistance. NAFLD refers to a spectrum of disorders ranging from asymptomatic hepatic steatosis (nonalcoholic fatty liver, NAFL) to nonalcoholic steatohepatitis (NASH), which increases the risk of developing more severe forms of liver disease such as progressive fibrosis, cirrhosis, and liver cancer. Currently, there are no food and drug administration (FDA) approved drugs to treat NASH. Pegbelfermin (BMS-986036) is a PEGylated fibroblast growth factor 21 (FGF21) analogue that is under investigation for the treatment of NASH.Areas covered: We reviewed the (pre)clinical pegbelfermin studies and compared these with other studies that assessed FGF21 and FGF21 analogues in the treatment of NASH.Expert opinion: With no FDA approved treatments available for NASH, there is an urgent need for novel therapies. Pegbelfermin is a systemic treatment with pleiotropic effects on various tissues. Short-term adverse effects are limited, but more research is required to study potential long-term safety issues. In a phase 2a trial, pegbelfermin has shown promising improvements in several NASH related outcomes. However, clinical trials demonstrating long-term benefits on hard outcomes such as liver histology, cirrhosis development, or survival are required for further validation.


Subject(s)
Fibroblast Growth Factors/analogs & derivatives , Non-alcoholic Fatty Liver Disease/drug therapy , Polyethylene Glycols/therapeutic use , Animals , Disease Progression , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/therapeutic use , Humans , Insulin Resistance , Non-alcoholic Fatty Liver Disease/physiopathology , Obesity/complications , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacology
4.
JAMA ; 322(14): 1360-1370, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31593273

ABSTRACT

Importance: Sprifermin is under investigation as a disease-modifying osteoarthritis drug. Objective: To evaluate the effects of sprifermin on changes in total femorotibial joint cartilage thickness in the more symptomatic knee of patients with osteoarthritis. Design, Setting, and Participants: FORWARD (FGF-18 Osteoarthritis Randomized Trial with Administration of Repeated Doses) was a 5-year, dose-finding, multicenter randomized clinical trial conducted at 10 sites. Eligible participants were aged 40 to 85 years with symptomatic, radiographic knee osteoarthritis and Kellgren-Lawrence grade 2 or 3. Enrollment began in July 2013 and ended in May 2014; the last participant visit occurred on May 8, 2017. The primary outcome at 2 years and a follow-up analysis at 3 years are reported. Interventions: Participants were randomized to 1 of 5 groups: intra-articular injections of 100 µg of sprifermin administered every 6 months (n = 110) or every 12 months (n = 110), 30 µg of sprifermin every 6 months (n = 111) or every 12 months (n = 110), or placebo every 6 months (n = 108). Each treatment consisted of weekly injections over 3 weeks. Main Outcomes and Measures: The primary end point was change in total femorotibial joint cartilage thickness measured by quantitative magnetic resonance imaging at 2 years. The secondary end points (of 15 total) included 2-year change from baseline in total Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores. The minimal clinically important difference (MCID) is unknown for the primary outcome; for total WOMAC score in patients with hip and knee osteoarthritis, the absolute MCID is 7 U (95% CI, 4 to 10 U) and the percentage MCID is 14% (95% CI, 9% to 18%). Results: Among 549 participants (median age, 65.0 years; 379 female [69.0%]), 474 (86.3%) completed 2-year follow-up. Compared with placebo, the changes from baseline to 2 years in total femorotibial joint cartilage thickness were 0.05 mm (95% CI, 0.03 to 0.07 mm) for 100 µg of sprifermin administered every 6 months; 0.04 mm (95% CI, 0.02 to 0.06 mm) for 100 µg of sprifermin every 12 months; 0.02 mm (95% CI, -0.01 to 0.04 mm) for 30 µg of sprifermin every 6 months; and 0.01 mm (95% CI, -0.01 to 0.03 mm) for 30 µg of sprifermin every 12 months. Compared with placebo, there were no statistically significant differences in mean absolute change from baseline in total WOMAC scores for 100 µg of sprifermin administered every 6 months or every 12 months, or for 30 µg of sprifermin every 6 months or every 12 months. The most frequently reported treatment-emergent adverse event was arthralgia (placebo: n = 46 [43.0%]; 100 µg of sprifermin administered every 6 months: n = 45 [41.3%]; 100 µg of sprifermin every 12 months: n = 50 [45.0%]; 30 µg of sprifermin every 6 months: n = 40 [36.0%]; and 30 µg of sprifermin every 12 months: n = 48 [44.0%]). Conclusions and Relevance: Among participants with symptomatic radiographic knee osteoarthritis, the intra-articular administration of 100 µg of sprifermin every 6 or 12 months vs placebo resulted in an improvement in total femorotibial joint cartilage thickness after 2 years that was statistically significant, but of uncertain clinical importance; there was no significant difference for 30 µg of sprifermin every 6 or 12 months vs placebo. Durability of response also was uncertain. Trial Registration: ClinicalTrials.gov Identifier: NCT01919164.


Subject(s)
Cartilage, Articular/drug effects , Fibroblast Growth Factors/administration & dosage , Osteoarthritis, Knee/drug therapy , Adult , Aged , Aged, 80 and over , Cartilage, Articular/pathology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Fibroblast Growth Factors/adverse effects , Follow-Up Studies , Humans , Injections, Intra-Articular , Knee Joint , Magnetic Resonance Imaging , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/pathology
5.
Am J Gastroenterol ; 113(5): 725-734, 2018 05.
Article in English | MEDLINE | ID: mdl-29717197

ABSTRACT

OBJECTIVE: NGM282 is an analog of fibroblast growth factor 19 (FGF19), a potent inhibitor of bile acid (BA) synthesis in animals and humans. In phase 2 trials in type 2 diabetes and primary biliary cholangitis, NGM282 was associated with dose-related abdominal cramping and diarrhea. We aimed to examine effects of NGM282 on colonic transit, stool frequency and consistency, hepatic BA synthesis (fasting serum C4), fecal fat, and BA in functional constipation (FC). METHODS: Two-dose NGM282 (1 and 6 mg, subcutaneously daily), parallel-group, randomized, placebo-controlled, 14-day study in patients with FC (Rome III criteria) and baseline colonic transit 24 h geometric center (GC) <3.0. We explored treatment interaction with SNPs in genes KLB, FGFR4, and TGR5 (GPBAR1). STATISTICAL ANALYSIS: overall ANCOVA at α = 0.025 (baseline as covariate where available), with three pairwise comparisons among the three groups (α = 0.008). RESULTS: Overall, NGM282 altered bowel function (number of bowel movements, looser stool form, and increased ease of passage) and significantly accelerated gastric and colonic transit. Dose-related effects were seen with GC 24 h, but not with gastric emptying (GE) and GC 48 h. There were no differences in fecal fat or weight, but there was reduced fecal total BA excretion with NGM282. The most common adverse events were increased appetite (n = 0 with placebo, 2 with 1 mg, 9 with 6 mg), injection site reaction (n = 2 placebo, 4 with 1 mg, 8 with 6 mg), and diarrhea (n = 1 with 1 mg and 4 with 6 mg NGM282). There was treatment interaction with KLB SNP, with greater increase in colonic transit in participants with the minor A allele (p = 0.056). CONCLUSION: NGM282 significantly impacts GE and colonic transit, consistent with the observed clinical symptoms. The specific mechanism of prokinetic activity requires further research.


Subject(s)
Constipation/drug therapy , Fibroblast Growth Factors/administration & dosage , Gastrointestinal Motility/drug effects , Gastrointestinal Transit/drug effects , Recombinant Proteins/administration & dosage , Adult , Appetite/drug effects , Bile Acids and Salts/biosynthesis , Constipation/genetics , Defecation/drug effects , Dose-Response Relationship, Drug , Feces/chemistry , Female , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/genetics , Humans , Injection Site Reaction/epidemiology , Injections, Subcutaneous , Klotho Proteins , Liver/drug effects , Liver/metabolism , Male , Membrane Proteins/genetics , Middle Aged , Polymorphism, Single Nucleotide , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptors, G-Protein-Coupled/genetics , Recombinant Proteins/adverse effects , Recombinant Proteins/genetics , Treatment Outcome
6.
Diabetes Obes Metab ; 19(12): 1762-1772, 2017 12.
Article in English | MEDLINE | ID: mdl-28573777

ABSTRACT

AIMS: To assess the safety, tolerability, pharmacokinetics and pharmacodynamics of PF-05231023, a long-acting fibroblast growth factor 21 (FGF21) analogue, in obese people with hypertriglyceridaemia on atorvastatin, with or without type 2 diabetes. METHODS: Participants received PF-05231023 or placebo intravenously once weekly for 4 weeks. Safety (12-lead ECGs, vital signs, adverse events [AEs], laboratory tests) and longitudinal weight assessments were performed. Blood samples were collected for pharmacokinetic and pharmacodynamic analyses. Cardiovascular safety studies were also conducted in telemetered rats and monkeys. Blood pressure (BP; mean, systolic and diastolic) and ECGs were monitored. RESULTS: A total of 107 people were randomized. PF-05231023 significantly decreased mean placebo-adjusted fasting triglycerides (day 25, 33%-43%) and increased HDL cholesterol (day 25, 15.7%-28.6%) and adiponectin (day 25, 1574 to 3272 ng/mL) across all doses, without significant changes in body weight (day 25, -0.45% to -1.21%). Modest decreases from baseline were observed for N-terminal propeptides of type 1 collagen (P1NP) on day 25, although C-telopeptide cross-linking of type 1 collagen (CTX-1) increased minimally. Systolic, diastolic BP, and pulse rate increased in a dose- and time-related manner. There were 5 serious AEs (one treatment-related) and no deaths. Three participants discontinued because of AEs. The majority of AEs were gastrointestinal. PF-05231023 increased BP and heart rate in rats, but not in monkeys. CONCLUSIONS: Once-weekly PF-05231023 lowered triglycerides markedly in the absence of weight loss, with modest changes in markers of bone homeostasis. This is the first report showing increases in BP and pulse rate in humans and rats after pharmacological administration of a long-acting FGF21 molecule.


Subject(s)
Anti-Obesity Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Bone Remodeling/drug effects , Fibroblast Growth Factors/therapeutic use , Hypertriglyceridemia/drug therapy , Hypolipidemic Agents/therapeutic use , Obesity/drug therapy , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/pharmacokinetics , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Biomarkers/blood , Body Mass Index , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/adverse effects , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Drug Resistance , Female , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/pharmacokinetics , Follow-Up Studies , Half-Life , Humans , Hypertension/chemically induced , Hypertension/physiopathology , Hypertriglyceridemia/blood , Hypertriglyceridemia/complications , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/adverse effects , Hypolipidemic Agents/pharmacokinetics , Infusions, Intravenous , Male , Middle Aged , Obesity/blood , Obesity/complications , Severity of Illness Index , Species Specificity
7.
Cochrane Database Syst Rev ; 6: CD011741, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28594443

ABSTRACT

BACKGROUND: Peripheral artery disease (PAD) is associated with a high clinical and socioeconomic burden. Treatments to alleviate the symptoms of PAD and decrease the risks of amputation and death are a high societal priority. A number of growth factors have shown a potential to stimulate angiogenesis. Growth factors delivered directly (as recombinant proteins), or indirectly (e.g. by viral vectors or DNA plasmids encoding these factors), have emerged as a promising strategy to treat patients with PAD. OBJECTIVES: To assess the effects of growth factors that promote angiogenesis for treating people with PAD of the lower extremities. SEARCH METHODS: The Cochrane Vascular Information Specialist searched the Specialised Register (June 2016) and CENTRAL (2016, Issue 5). We searched trial registries for details of ongoing or unpublished studies. We also checked the reference lists of relevant publications and, if necessary, tried to contact the trialists for details of the studies. SELECTION CRITERIA: We included randomised controlled trials comparing growth factors (delivered directly or indirectly) with no intervention, placebo or any other intervention not based on the growth factor's action in patients with PAD of the lower extremities. The primary outcomes were limb amputation, death and adverse events. The secondary outcomes comprised walking ability, haemodynamic measures, ulceration and rest pain. DATA COLLECTION AND ANALYSIS: Two review authors independently selected trials and assessed the risk of bias. We used outcomes of the studies at low risk of bias for the main analysis and of all studies in the sensitivity analyses. We calculated odds ratios (OR) for dichotomous outcomes and mean differences for continuous outcomes with 95% confidence intervals (CI). We evaluated statistical heterogeneity using the I2 statistic and Cochrane's Q test. We conducted meta-analysis for the overall effect and for each growth factor as a subgroup analysis using OR in a fixed-effect model. We evaluated the robustness of the results in a sensitivity analysis using risk ratio (RR) and/or a random-effects model. We also assessed the quality of the evidence for each outcome. MAIN RESULTS: We included 20 trials in the review and used 14 studies (on approximately 1400 participants) with published results in the analyses. Six published studies compared fibroblast growth factors (FGF), four studies hepatocyte growth factors (HGF) and another four studies vascular endothelial growth factors (VEGF), versus placebo or no therapy. Six of these studies exclusively or mainly investigated participants with intermittent claudication and eight studies exclusively participants with critical limb ischaemia. Follow-up generally ranged from three months to one year. Two small studies provided some data at 2 years and one of them also at 10 years.The direction and size of effects for growth factors on major limb amputations (OR 0.99, 95% CI 0.71 to 1.38; 10 studies, N = 1075) and death (OR 0.99, 95% CI 0.69 to 1.41; 12 studies, N = 1371) at up to two years are uncertain. The quality of the evidence is low due to risk of bias and imprecision (at one year, moderate-quality evidence due to imprecision). However, growth factors may decrease the rate of any limb amputations (OR 0.56, 95% CI 0.31 to 0.99; 6 studies, N = 415). The quality of the evidence is low due to risk of bias and selective reporting.The direction and size of effects for growth factors on serious adverse events (OR 1.09, 95% CI 0.79 to 1.50; 13 studies, N = 1411) and on any adverse events (OR 1.10, 95% CI 0.73 to 1.64; 4 studies, N = 709) at up to two years are also uncertain. The quality of the evidence is low due to risk of bias and imprecision (for serious adverse events at one year, moderate-quality evidence due to imprecision).Growth factors may improve haemodynamic measures (low-quality evidence), ulceration (very low-quality evidence) and rest pain (very low-quality evidence) up to one year, but they have little or no effect on walking ability (low-quality evidence). We did not identify any relevant differences in effects between growth factors (FGF, HGF and VEGF). AUTHORS' CONCLUSIONS: The results of this review do not support the use of therapy with the growth factors FGF, HGF or VEGF in people with PAD of the lower extremities to prevent death or major limb amputation or to improve walking ability. However, the use of these growth factors may improve haemodynamic measures and decrease the rate of any limb amputations (probably due to preventing minor amputations) with an uncertain effect on adverse events; an improvement of ulceration and rest pain is very uncertain. New trials at low risk of bias are needed to generate evidence with more certainty.


Subject(s)
Fibroblast Growth Factors/therapeutic use , Hepatocyte Growth Factor/therapeutic use , Intermittent Claudication/drug therapy , Peripheral Arterial Disease/drug therapy , Vascular Endothelial Growth Factor A/therapeutic use , Amputation, Surgical/statistics & numerical data , Fibroblast Growth Factors/adverse effects , Hepatocyte Growth Factor/adverse effects , Humans , Intermittent Claudication/mortality , Leg/blood supply , Leg/surgery , Leg Ulcer/drug therapy , Peripheral Arterial Disease/mortality , Randomized Controlled Trials as Topic , Vascular Endothelial Growth Factor A/adverse effects
8.
J Diabetes Complications ; 31(4): 726-734, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28143735

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a promising regulator of glucose and lipid metabolism with multiple beneficial effects including hypoglycemic and lipid-lowering. Previous studies have reported that FGF21 is expected to become a new drug for treatment of diabetes. Liraglutide and insulin glargine are the two representative anti-diabetic biological drugs. In the current study, we aim to compare the long-term pharmacological efficacy of mFGF21 (an FGF21 analogue), liraglutide and insulin glargine in type 2 diabetic db/db mice. Db/db mice were initially treated with three kinds of proteins (25nmol/kg/day) by subcutaneous injection once a day for 4weeks, then subsequently be treated with once every two days for next 4weeks. After 8weeks of treatments, the blood glucose levels, body weights, glycosylated hemoglobin levels, fasting insulin levels, serum lipid profiles, hepatic biochemical parameters, oral glucose tolerance tests and hepatic mRNA expression levels of several proteins (GK, G6P, GLUT-1 and GLUT-4) associated with glucose metabolism of the experimental mice were detected. Results demonstrated that three proteins could significantly decrease the fed blood glucose levels of db/db mice. After treatment for 1week, the fed blood glucose levels of db/db mice in liraglutide group were significantly lower than those in mFGF21 and insulin glargine groups. However, after 2weeks of administration, the long-lasting hypoglycemic effect of mFGF21 was superior to liraglutide and insulin glargine up to the end of the experiments. Compared with liraglutide and insulin glargine, mFGF21 significantly reduced the glycosylated hemoglobin levels and improved the ability on glycemic control, insulin resistance, serum lipid and liver function states in db/db mice after 8weeks treatments. In addition, mFGF21 regulated glucose metabolism through increasing the mRNA expression levels of GK and GLUT-1, and decreasing the mRNA expression level of G6P. But liraglutide and insulin glargine could only up-regulate the mRNA expression of GLUT-4. In summary, as a hypoglycemic drug for long-term treatment, mFGF21 has the potential to be an ideal drug candidate for the therapy of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drugs, Investigational/therapeutic use , Fibroblast Growth Factors/therapeutic use , Hypoglycemic Agents/therapeutic use , Insulin Glargine/therapeutic use , Liraglutide/therapeutic use , Mutant Proteins/therapeutic use , Animals , Biomarkers/blood , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Drugs, Investigational/adverse effects , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacology , Endopeptidases/metabolism , Female , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hyperglycemia/prevention & control , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Insulin Glargine/adverse effects , Insulin Glargine/pharmacology , Insulin Resistance , Liraglutide/adverse effects , Liraglutide/pharmacology , Mice, Inbred C57BL , Mice, Mutant Strains , Mutant Proteins/adverse effects , Mutant Proteins/genetics , Mutant Proteins/pharmacology , Random Allocation , Recombinant Proteins/adverse effects , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
9.
PLoS One ; 12(2): e0171703, 2017.
Article in English | MEDLINE | ID: mdl-28207794

ABSTRACT

We evaluated the mitigating effects of fibroblast growth factor 4 and 7 (FGF4 and FGF7, respectively) in comparison with long acting protected graft copolymer (PGC)-formulated FGF4 and 7 (PF4 and PF7, respectively) administered to C57BL/6J mice a day after exposure to LD50/30 (15.7 Gy) partial body irradiation (PBI) which targeted the gastrointestinal (GI) system. The PGC that we developed increased the bioavailability of FGF4 and FGF7 by 5- and 250-fold compared to without PGC, respectively, and also sustained a 24 hr presence in the blood after a single subcutaneous administration. The dose levels tested for mitigating effects on radiation injury were 3 mg/kg for the PF4 and PF7 and 1.5 mg each for their combination (PF4/7). Amifostine administered prior to PBI was used as a positive control. The PF4, PF7, or PF4/7 mitigated the radiation lethality in mice. The mitigating effect of PF4 and PF7 was similar to the positive control and PF7 was better than other mitigators tested. The plasma citrulline levels and hematology parameters were early markers of recovery and survival. GI permeability function appeared to be a late or full recovery indicator. The villus length and crypt number correlated with plasma citrulline level, indicating that it can act as a surrogate marker for these histology evaluations. The IL-18 concentrations in jejunum as early as day 4 and TPO levels in colon on day 10 following PBI showed statistically significant changes in irradiated versus non-irradiated mice which makes them potential biomarkers of radiation exposure. Other colon and jejunum cytokine levels are potentially useful but require larger numbers of samples than in the present study before their full utility can be realized.


Subject(s)
Acute Radiation Syndrome/drug therapy , Fibroblast Growth Factors/therapeutic use , Gastrointestinal Tract/drug effects , Radiation Injuries, Experimental/therapy , Animals , Cell Proliferation/drug effects , Drug Delivery Systems/methods , Female , Fibroblast Growth Factor 4/adverse effects , Fibroblast Growth Factor 4/therapeutic use , Fibroblast Growth Factor 7/adverse effects , Fibroblast Growth Factor 7/therapeutic use , Fibroblast Growth Factors/adverse effects , Fibroblasts/drug effects , Gastrointestinal Tract/pathology , Gastrointestinal Tract/radiation effects , Kaplan-Meier Estimate , Lethal Dose 50 , Male , Mice, Inbred C57BL , Polylysine/chemistry , Polymers/chemistry
10.
J Intern Med ; 281(3): 233-246, 2017 03.
Article in English | MEDLINE | ID: mdl-27878865

ABSTRACT

Fibroblast growth factor (FGF) 21 belongs to a hormone-like subgroup within the FGF superfamily. The members of this subfamily, FGF19, FGF21 and FGF23, are characterized by their reduced binding affinity for heparin that enables them to be transported in the circulation and function in an endocrine manner. It is likely that FGF21 also acts in an autocrine and paracrine fashion, as multiple organs can produce this protein and its plasma concentration seems to be below the level necessary to induce a pharmacological effect. FGF21 signals via FGF receptors, but for efficient receptor engagement it requires a cofactor, membrane-spanning ßKlotho (KLB). The regulation of glucose uptake in adipocytes was the initial biological activity ascribed to FGF21, but this hormone is now recognized to stimulate many other pathways in vitro and display multiple pharmacological effects in metabolically compromised animals and humans. Understanding of the precise physiology of FGF21 and its potential medicinal role has evolved exponentially over the last decade, yet numerous aspects remain to be defined and others are a source of debate. Here we provide a historical overview of the advances in FGF21 biology focusing on the uncertainties in the mechanism of action as well as the differing viewpoints relating to this intriguing protein.


Subject(s)
Fibroblast Growth Factors/physiology , Adipocytes/metabolism , Animals , Blood Glucose/metabolism , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/pharmacology , Glucose/metabolism , Humans , Lipolysis , Obesity/physiopathology , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction
11.
Clin Exp Rheumatol ; 34(3): 445-50, 2016.
Article in English | MEDLINE | ID: mdl-27050139

ABSTRACT

OBJECTIVES: To evaluate the safety of intra-articular sprifermin (primary), and to evaluate systemic exposure, biomarkers, histology, and other cartilage parameters in patients with advanced osteoarthritis (OA). METHODS: This was a first-in-human, double-blind, randomised, placebo-controlled trial of single and multiple ascending doses of sprifermin from 3-300 µg in knee OA patients scheduled for total knee replacement. Patients were randomised 3:1 to sprifermin or placebo, injected into the target knee once or once weekly for 3 weeks, and followed-up for 24 weeks. RESULTS: Fifty-five patients were treated with sprifermin, 25 with single and 30 with multiple doses, 18 received placebo. There was no clear difference between the active and placebo groups in incidence, severity, and nature of reported treatment emergent adverse events. Acute inflammatory reactions were slightly more common with sprifermin 300 µg, but none led to discontinuation. No clear difference was seen between placebo and sprifermin in physician-assessed local tolerability, pain, or swelling in the knee. No meaningful changes over time, or differences between treatment groups, were observed for safety laboratory parameters or ECG. Although individual abnormalities were observed, no patterns were evident suggesting a relation to treatment or potential safety concern. No systemic sprifermin exposure, anti-FGF18 antibodies, or clear-cut effects on systemic biomarkers were detected. CONCLUSIONS: This first clinical trial of sprifermin revealed no serious safety concerns, although larger studies are needed. The possibility of positive effects of intra-articular sprifermin on histological and other cartilage parameters in knee OA also warrant further investigation.


Subject(s)
Cartilage, Articular , Fibroblast Growth Factors , Osteoarthritis, Knee , Adult , Aged , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Monitoring/methods , Female , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/adverse effects , Growth Substances , Humans , Injections, Intra-Articular , Knee Joint/pathology , Male , Middle Aged , Osteoarthritis, Knee/diagnosis , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/physiopathology , Patient Acuity , Treatment Outcome
13.
Br J Clin Pharmacol ; 80(5): 1051-63, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25940675

ABSTRACT

AIMS: The aim of the present study was to evaluate the pharmacokinetics/pharmacodynamics (PK/PD), safety and tolerability of single intravenous (IV) doses of PF-05231023, a long acting fibroblast growth factor 21 (FGF21) analogue being developed for the treatment of type 2 diabetes mellitus (T2DM). METHODS: T2DM subjects (glycosylated haemoglobin: 7.0-10.5%; on stable metformin therapy and/or diet and exercise) were randomized to receive a single dose of placebo or PF-05231023 (0.5-200 mg). Safety evaluations were performed up to 14 days after dosing. PK and PD endpoints were measured and a PK/PD model was developed for triglyceride - an early marker of drug activity. RESULTS: No antidrug antibody or serious adverse events (AEs) were observed. The most frequent AEs were gastrointestinal but were generally mild. Plasma PF-05231023 levels peaked immediately post-IV dosing, with mean terminal half-lives of 6.5-7.7 h and 66.5- 96.6 h for intact C- and N-termini, respectively. Intact C-terminus exposures increased proportionally with increasing dose, whereas N-terminus exposures appeared to trend higher than dose-proportionally. Although no apparent effect on plasma glucose was seen, dose-dependent decreases in triglyceride were observed, with a maximum reduction of 48.5 ± 10.0% (mean ± standard deviation) for the 200 mg dose compared with a reduction of 19.1 ± 26.4% for placebo, demonstrating proof of pharmacology. Moreover, a reduction in total cholesterol and low-density lipoprotein cholesterol and an increase in high-density lipoprotein cholesterol were observed in the high-dose groups. CONCLUSIONS: Single IV doses of PF-05231023 up to 200 mg were generally safe and well tolerated by subjects with T2DM. The observed early sign of pharmacology supports further clinical testing of PF-05231023 upon repeated administration.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Diabetes Mellitus, Type 2/blood , Fibroblast Growth Factors/agonists , Fibroblast Growth Factors/pharmacokinetics , Administration, Intravenous , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Dose-Response Relationship, Drug , Female , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/pharmacology , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Male , Middle Aged , Triglycerides/blood
14.
Int Immunopharmacol ; 25(1): 74-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25601498

ABSTRACT

It has been demonstrated that circulating FGF21 levels are elevated in the serum and synovial fluid of patients with rheumatoid arthritis (RA). The aim of this study is to investigate efficacy of FGF21 for treatment of RA and the molecular mechanisms of the therapeutic effect on collagen-induced arthritis (CIA). Mice with CIA were subcutaneously administered with FGF21 (5, 2 or 1mg·kg(-1)·d(-1)), IL-1ß antibody (5mg·kg(-1)·d(-1)), IL-17A antibody (5mg·kg(-1)·d(-1)) and dexamethasone (DEX) (1mg·kg(-1)·d(-1)), respectively. The effects of treatment were determined by arthritis severity score, histological damage and cytokine production. The activation of NF-κB was analyzed by Western blotting. We also detected the levels of oxidative stress parameters. Our results showed that FGF21 had beneficial effects on clinical symptom and histological lesion of CIA mice. Similar to antibody and DEX, FGF21 treatment alleviated the severity of arthritis by reducing humoral and cellular immune responses and down-regulating the expression of pro-inflammatory cytokines. FGF21 treatment also reduced the expression of TNF-α, IL-1ß, IL-6, IFN-γ and MMP-3 and increased level of IL-10 in the spleen tissue or the plasma of CIA mice in a dose-dependent manner. Furthermore, FGF21 inhibited IκBα degradation and NF-κB p65 nuclear translocation and induced significant changes of oxidative stress parameters (MDA, SOD, CAT, GSH-PX and GSH) in the plasma. FGF21 exerts therapeutic efficacy for RA through antioxidant reaction and inhibiting NF-κB inflammatory pathway. This study provides evidence that FGF21 may be a promising therapeutic agent for RA patients.


Subject(s)
Antioxidants/administration & dosage , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Fibroblast Growth Factors/administration & dosage , NF-kappa B/metabolism , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/adverse effects , Antioxidants/adverse effects , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dexamethasone/administration & dosage , Dexamethasone/adverse effects , Disease Progression , Fibroblast Growth Factors/adverse effects , Humans , Immunosuppression Therapy , Inflammation Mediators/metabolism , Male , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred DBA , Oxidative Stress/drug effects , Signal Transduction/drug effects
15.
Sci Transl Med ; 6(247): 247ra100, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25080475

ABSTRACT

Hepatic accumulation of bile acids is central to the pathogenesis of cholestatic liver diseases. Endocrine hormone fibroblast growth factor 19 (FGF19) may reduce hepatic bile acid levels through modulation of bile acid synthesis and prevent subsequent liver damage. However, FGF19 has also been implicated in hepatocellular carcinogenesis, and consequently, the potential risk from prolonged exposure to supraphysiological levels of the hormone represents a major hurdle for developing an FGF19-based therapy. We describe a nontumorigenic FGF19 variant, M70, which regulates bile acid metabolism and, through inhibition of bile acid synthesis and reduction of excess hepatic bile acid accumulation, protects mice from liver injury induced by either extrahepatic or intrahepatic cholestasis. Administration of M70 in healthy human volunteers potently reduces serum levels of 7α-hydroxy-4-cholesten-3-one, a surrogate marker for the hepatic activity of cholesterol 7α-hydroxylase (CYP7A1), the enzyme responsible for catalyzing the first and rate-limiting step in the classical bile acid synthetic pathway. This study provides direct evidence for the regulation of bile acid metabolism by FGF19 pathway in humans. On the basis of these results, the development of nontumorigenic FGF19 variants capable of modulating CYP7A1 expression represents an effective approach for the prevention and treatment of cholestatic liver diseases as well as potentially for other disorders associated with bile acid dysregulation.


Subject(s)
Bile Acids and Salts/metabolism , Cholagogues and Choleretics/therapeutic use , Cholestasis, Extrahepatic/drug therapy , Cholestasis, Intrahepatic/drug therapy , Fibroblast Growth Factors/therapeutic use , Liver/drug effects , Adult , Animals , Australia , Biomarkers/blood , Cholagogues and Choleretics/adverse effects , Cholagogues and Choleretics/pharmacokinetics , Cholestasis, Extrahepatic/genetics , Cholestasis, Extrahepatic/metabolism , Cholestasis, Extrahepatic/pathology , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/pathology , Cholestenones/blood , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Disease Models, Animal , Double-Blind Method , Down-Regulation , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacokinetics , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques , Genetic Variation , Healthy Volunteers , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Middle Aged , RNA, Messenger/metabolism , Recombinant Proteins/therapeutic use , Risk Assessment , Young Adult
16.
Biol Pharm Bull ; 37(7): 1149-57, 2014.
Article in English | MEDLINE | ID: mdl-24989006

ABSTRACT

Disabilities triggered by neurodegeneration mainly result in mortality in the elderly, and patients with neurodegenerative disease also display deficits in olfactory function. Therefore drug distribution to the brain through intranasal administration has become one of the most difficult challenges in the treatment of central nervous system (CNS) diseases. TAT-human acidic fibroblast growth factor (HaFGF) is a new fused protein retaining the neuroprotective activities of HaFGF, and is a promising prospect in the treatment of neurodegenerative diseases. TAT (a cell-penetrating peptide) contains a high relative abundance of positively charged amino acids such as lysine and arginine, which have a powerful attraction to the negatively charge on the nasal epithelial membrane. The present study focused on the evaluation of the safety and absorption characteristics of TAT-HaFGF following intranasal administration. After TAT-HaFGF intranasal administration (100, 300, 600 µg/kg) for 5 weeks, hematoxylin-eosin (HE) staining showed no pathology in any of the investigated tissues and organs. The expression of olfactory marker protein (OMP) was observed with immunohistochemical staining, which showed no altered expression in the sensory neurons of the nasal epithelium. Nasal ciliotoxicity studies carried out using an in situ palate model and optical microscope showed that TAT-HaFGF had no nasal ciliotoxicity. The distribution of the TAT-HaFGF following intranasal administration was assessed using a radioisotopic tracing method. Radioactivity was observed in the brain after 15 min. This became stronger at 30 min and weaker at 1 h. All of the results confirmed the in vivo safety of TAT-HaFGF via intranasal administration.


Subject(s)
Brain/drug effects , Fibroblast Growth Factors/administration & dosage , Gene Products, tat/administration & dosage , Nasal Absorption/drug effects , Nasal Mucosa/drug effects , Neuroprotective Agents/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Administration, Intranasal , Animals , Brain/metabolism , Bufo bufo , Cilia/drug effects , Female , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/pharmacokinetics , Gene Products, tat/adverse effects , Gene Products, tat/pharmacokinetics , Male , Nasal Mucosa/metabolism , Neuroprotective Agents/adverse effects , Neuroprotective Agents/pharmacokinetics , Olfactory Bulb/drug effects , Olfactory Bulb/metabolism , Rats, Sprague-Dawley , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Tissue Distribution
17.
Arthritis Rheumatol ; 66(7): 1820-31, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24740822

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of intraarticular sprifermin (recombinant human fibroblast growth factor 18) in the treatment of symptomatic knee osteoarthritis (OA). METHODS: The study was a randomized, double-blind, placebo-controlled, proof-of-concept trial. Intraarticular sprifermin was evaluated at doses of 10 µg, 30 µg, and 100 µg. The primary efficacy end point was change in central medial femorotibial compartment cartilage thickness at 6 months and 12 months as determined using quantitative magnetic resonance imaging (qMRI). The primary safety end points were nature, incidence, and severity of local and systemic treatment-emergent adverse events (AEs) and acute inflammatory reactions, as well as results of laboratory assessments. Secondary end points included changes in total and compartment femorotibial cartilage thickness and volume as assessed by qMRI, changes in joint space width (JSW) seen on radiographs, and pain scores on the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC). RESULTS: One hundred ninety-two patients were randomized and evaluated for safety, 180 completed the trial, and 168 were evaluated for the primary efficacy end point. We found no statistically significant dose response in change in central medial femorotibial compartment cartilage thickness. Sprifermin was associated with statistically significant, dose-dependent reductions in loss of total and lateral femorotibial cartilage thickness and volume and in JSW narrowing in the lateral femorotibial compartment. All groups had improved WOMAC pain scores, with statistically significantly less improvement at 12 months in patients receiving the 100-µg dose of sprifermin as compared with those receiving placebo. There was no significant difference in serious AEs, treatment-emergent AEs, or acute inflammatory reactions between sprifermin and placebo groups. CONCLUSION: No statistically significant relationship between treatment group and reduction in central medial femorotibial compartment cartilage thickness was observed; however, prespecified structural secondary end points showed statistically significant dose-dependent reductions after sprifermin treatment. Sprifermin was not associated with any local or systemic safety concerns.


Subject(s)
Fibroblast Growth Factors/administration & dosage , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/immunology , Aged , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Double-Blind Method , Female , Fibroblast Growth Factors/adverse effects , Humans , Injections, Intra-Articular , Knee Joint/drug effects , Knee Joint/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Osteoarthritis, Knee/pathology , Placebos , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Treatment Outcome
18.
Endocrinology ; 155(5): 2031-40, 2014 May.
Article in English | MEDLINE | ID: mdl-24564398

ABSTRACT

Thyroid hormone (TH) regulates fibroblast growth factor 21 (FGF21) levels in the liver and in the adipose tissue. In contrast, peripheral FGF21 administration leads to decreased circulating levels of TH. These data suggest that FGF21 and TH could interact to regulate metabolism. In the present study, we confirmed that TH regulates adipose and hepatic FGF21 expression and serum levels in mice. We next investigated the influence of TH administration on key serum metabolites, gene expression in the liver and brown adipose tissue, and energy expenditure in FGF21 knockout mice. Surprisingly, we did not observe any significant differences in the effects of TH on FGF21 knockout mice compared with those in wild-type animals, indicating that TH acts independently of FGF21 for the specific outcomes studied. Furthermore, exogenous FGF21 administration to hypothyroid mice led to similar changes in serum and liver lipid metabolites and gene expression in both hypothyroid and euthyroid mice. Thus, it appears that FGF21 and TH have similar actions to decrease serum and liver lipids despite having some divergent regulatory effects. Whereas TH leads to up-regulation in the liver and down-regulation in brown adipose tissue of genes involved in the lipid synthesis pathway (eg, fatty acid synthase (FASN) and SPOT14), FGF21 leads to the opposite changes in expression of these genes. In conclusion, TH and FGF21 act independently on the outcomes studied, despite their ability to regulate each other's circulating levels. Thus, TH and FGF21 may modulate the availability of each other in critical metabolic states.


Subject(s)
Fibroblast Growth Factors/therapeutic use , Gene Expression Regulation/drug effects , Hypothyroidism/drug therapy , Lipid Metabolism/drug effects , Lipotropic Agents/therapeutic use , Thyroxine/therapeutic use , Triiodothyronine/therapeutic use , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Animals , Energy Metabolism/drug effects , Female , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hormone Replacement Therapy/adverse effects , Hyperlipidemias/etiology , Hyperlipidemias/prevention & control , Hyperthyroidism/blood , Hyperthyroidism/chemically induced , Hyperthyroidism/metabolism , Hypothyroidism/blood , Hypothyroidism/metabolism , Hypothyroidism/physiopathology , Lipotropic Agents/adverse effects , Lipotropic Agents/blood , Lipotropic Agents/poisoning , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Thyroxine/adverse effects , Thyroxine/blood , Thyroxine/poisoning , Triiodothyronine/adverse effects , Triiodothyronine/blood
19.
Support Care Cancer ; 21(1): 343-55, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22987094

ABSTRACT

PURPOSE: The aim of this project was to review the literature and define clinical practice guidelines for the use of cytokines and growth factor agents for the prevention or treatment of oral mucositis induced by cancer chemotherapy or radiotherapy. METHODS: A systematic review was conducted by the Mucositis Study Group of the Multinational Association of Supportive Care in Cancer/International Society for Oral Oncology (MASCC/ISOO). The body of evidence for each intervention, in each cancer treatment setting, was assigned an evidence level. Based on the evidence level, one of the following three guideline determinations was possible: Recommendation, Suggestion, No guideline possible. RESULTS: Sixty-four clinical studies across 11 interventions were evaluated. A recommendation was made for the use of recombinant human KGF-1 (palifermin) at a dose of 60 µg/kg per day for 3 days prior to conditioning treatment and for 3 days post-transplant for prevention of oral mucositis in patients receiving high-dose chemotherapy and total body irradiation followed by autologous stem cell transplantation for hematological malignancies. A suggestion was made against using granulocyte macrophage colony-stimulating factor mouthwash for the prevention of oral mucositis in the setting of high-dose chemotherapy followed by autologous or allogeneic stem cell transplantation. No guideline was possible for any other cytokine or growth factor agents due to inconclusive evidence. CONCLUSIONS: Of the cytokine and growth factor agents studied for oral mucositis, the evidence only supports use of palifermin in the specific population listed above. Additional well-designed research is needed on other cytokine and growth factor interventions and in other cancer treatment settings.


Subject(s)
Cytokines/therapeutic use , Intercellular Signaling Peptides and Proteins/therapeutic use , Neoplasms/complications , Stomatitis/therapy , Cytokines/adverse effects , Evidence-Based Medicine , Fibroblast Growth Factor 7/adverse effects , Fibroblast Growth Factor 7/therapeutic use , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/therapeutic use , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Intercellular Signaling Peptides and Proteins/adverse effects , Mouthwashes , Neoplasms/drug therapy , Neoplasms/radiotherapy , Practice Guidelines as Topic , Stomatitis/etiology , Stomatitis/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...