Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Nutr J ; 12: 160, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24330605

ABSTRACT

BACKGROUND: Lactose intolerance (LI) is a common medical problem with limited treatment options. The primary symptoms are abdominal pain, diarrhea, bloating, flatulence, and cramping. Limiting dairy foods to reduce symptoms contributes to low calcium intake and the risk for chronic disease. Adaptation of the colon bacteria to effectively metabolize lactose is a novel and potentially useful approach to improve lactose digestion and tolerance. RP-G28 is novel galacto-oligosaccharide (GOS) being investigated to improve lactose digestion and the symptoms of lactose intolerance in affected patients. METHODS: A randomized, double-blind, parallel group, placebo-controlled study was conducted at 2 sites in the United States. RP-G28 or placebo was administered to 85 patients with LI for 35 days. Post-treatment, subjects reintroduced dairy into their daily diets and were followed for 30 additional days to evaluate lactose digestion as measured by hydrogen production and symptom improvements via a patient-reported symptom assessment instrument. RESULTS: Lactose digestion and symptoms of LI trended toward improvement on RP-G28 at the end of treatment and 30 days post-treatment. A reduction in abdominal pain was also demonstrated in the study results. Fifty percent of RP-G28 subjects with abdominal pain at baseline reported no abdominal pain at the end of treatment and 30 days post treatment (p = 0.0190). RP-G28 subjects were also six times more likely to claim lactose tolerance post-treatment once dairy foods had been re-introduced into their diets (p = 0.0389). CONCLUSIONS: Efficacy trends and favorable safety/tolerability findings suggest that RP-G28 appears to be a potentially useful approach for improving lactose digestion and LI symptoms. The concurrent reduction in abdominal pain and improved overall tolerance could be a meaningful benefit to lactose intolerant individuals.


Subject(s)
Digestion , Gastrointestinal Agents/therapeutic use , Lactase/deficiency , Lactose Intolerance/diet therapy , Lactose/metabolism , Oligosaccharides/therapeutic use , Prebiotics , Abdominal Pain/epidemiology , Abdominal Pain/etiology , Abdominal Pain/prevention & control , Adult , Colon/microbiology , Dairy Products/adverse effects , Double-Blind Method , Female , Follow-Up Studies , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/adverse effects , Humans , Incidence , Intestinal Mucosa/microbiology , Lactose Intolerance/microbiology , Lactose Intolerance/physiopathology , Male , Oligosaccharides/administration & dosage , Oligosaccharides/adverse effects , Prebiotics/adverse effects , Prebiotics/analysis , Severity of Illness Index , Trisaccharides/administration & dosage , Trisaccharides/adverse effects , Trisaccharides/therapeutic use , United States/epidemiology
2.
Stem Cell Res Ther ; 3(4): 26, 2012 Jul 09.
Article in English | MEDLINE | ID: mdl-22776246

ABSTRACT

Aastrom Biosciences has developed a proprietary cell-processing technology that enables the manufacture of ixmyelocel-T, a patient-specific multicellular therapy expanded from a small sample of a patient's own bone marrow. Ixmyelocel-T is produced under current good manufacturing practices (cGMP) in a fully closed, automated system that expands mesenchymal stem cells (MSCs) and macrophages. While the cell types in ixmyelocel-T are the same as those found in the bone marrow, the numbers of MSCs and alternative macrophages are greater in ixmyelocel-T. We propose that the mixture of expanded MSCs and alternatively activated macrophages promote long-term tissue repair of ischemic tissue. The multiple cell types in ixmyelocel-T have a range of biological activities that are likely to contribute to a complex mechanism of action. Clinical trial data collected to date support the potential for ixmyelocel-T as an efficacious and safe treatment for ischemic cardiovascular indications, including critical limb ischemia (CLI) and a severe form of heart failure, dilated cardiomyopathy (DCM). The CLI clinical program has completed phase 2 and has reached concurrence with the Food and Drug Administration (FDA) on a phase 3 study (REVIVE) through the Special Protocol Assessment (SPA) process. The phase 3 study began screening patients in February 2012. The DCM clinical program will initiate phase 2b in 2012.


Subject(s)
Drug Industry , Antigens, CD/metabolism , Cardiovascular Diseases/mortality , Cardiovascular Diseases/therapy , Clinical Trials as Topic , Humans , Kaplan-Meier Estimate , Macrophages/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phenotype
3.
Mol Ther ; 20(6): 1280-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22453769

ABSTRACT

Ixmyelocel-T is a patient-specific, expanded, multicellular therapy evaluated in patients with lower extremity critical limb ischemia (CLI) with no options for revascularization. This randomized, double-blind, placebo-controlled, phase 2 trial (RESTORE-CLI) compared the efficacy and safety of intramuscular injections of ixmyelocel-T with placebo. Patients received one-time injections over 20 locations in a single leg and were followed for 12 months. Safety assessments included occurrence of adverse events. Efficacy assessments included time to first occurrence of treatment failure (TTF; major amputation of injected leg; all-cause mortality; doubling of total wound surface area from baseline; de novo gangrene) and amputation-free survival (AFS; major amputation of injected leg; all-cause mortality). A total of 77 patients underwent bone marrow or sham aspiration; 72 patients received ixmyelocel-T (48 patients) or placebo (24 patients). Adverse event rates were similar. Ixmyelocel-T treatment led to a significantly prolonged TTF (P = 0.0032, logrank test). AFS had a clinically meaningful 32% reduction in event rate that was not statistically significant (P = 0.3880, logrank test). Treatment effect in post hoc analyses of patients with baseline wounds was more pronounced (TTF: P < 0.0001, AFS: P = 0.0802, logrank test). Ixmyelocel-T treatment was well tolerated and may offer a potential new treatment option.


Subject(s)
Ischemia/therapy , Lower Extremity/blood supply , Macrophages/transplantation , Mesenchymal Stem Cell Transplantation , Aged , Aged, 80 and over , Amputation, Surgical , Cell Transplantation/adverse effects , Female , Humans , Ischemia/mortality , Ischemia/surgery , Male , Middle Aged , Survival Analysis , Treatment Failure , Treatment Outcome
4.
J Vasc Surg ; 54(4): 1032-41, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21684715

ABSTRACT

OBJECTIVES: Cell therapy is a novel experimental treatment modality for patients with critical limb ischemia (CLI) of the lower extremities and no other established treatment options. This study was conducted to assess the safety and clinical efficacy of intramuscular injection of autologous tissue repair cells (TRCs). METHODS: A prospective, randomized double-blinded, placebo controlled, multicenter study (RESTORE-CLI) was conducted at 18 centers in the United States in patients with CLI and no option for revascularization. Enrollment of 86 patients began in April 2007 and ended in February 2010. For the prospectively planned interim analysis, conducted in February 2010, 33 patients had the opportunity to complete the trial (12 months of follow-up), and 46 patients had completed at least 6 months of follow-up. The interim analysis included analysis of both patient populations. An independent physician performed the bone marrow or sham control aspiration. The aspirate was processed in a closed, automated cell manufacturing system for approximately 12 days to generate the TRC population of stem and progenitor cells. An average of 136 ± 41 × 10(6) total viable cells or electrolyte (control) solution were injected into 20 sites in the ischemic lower extremity. The primary end point was safety as evaluated by adverse events, and serious adverse events as assessed at multiple follow-up time points. Clinical efficacy end points included major amputation-free survival and time to first occurrence of treatment failure (defined as any of the following: major amputation, death, de novo gangrene, or doubling of wound size), as well as major amputation rate and measures of wound healing. RESULTS: There was no difference in adverse or serious adverse events between the two groups. Statistical analysis revealed a significant increase in time to treatment failure (log-rank test, P = .0053) and amputation-free survival in patients receiving TRC treatment, (log-rank test, P = .038). Major amputation occurred in 19% of TRC-treated patients compared to 43% of controls (P = .14, Fisher exact test). There was evidence of improved wound healing in the TRC-treated patients when compared with controls at 12 months. CONCLUSIONS: Intramuscular injection of autologous bone marrow-derived TRCs is safe and decreases the occurrence of clinical events associated with disease progression when compared to placebo in patients with lower extremity CLI and no revascularization options.


Subject(s)
Bone Marrow Transplantation , Ischemia/surgery , Lower Extremity/blood supply , Stem Cell Transplantation , Aged , Aged, 80 and over , Amputation, Surgical , Bone Marrow Transplantation/adverse effects , Bone Marrow Transplantation/mortality , Cells, Cultured , Double-Blind Method , Female , Humans , Injections, Intramuscular , Ischemia/mortality , Ischemia/pathology , Ischemia/physiopathology , Kaplan-Meier Estimate , Limb Salvage , Male , Middle Aged , Prospective Studies , Reoperation , Risk Assessment , Risk Factors , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/mortality , Time Factors , Transplantation, Autologous , Treatment Outcome , United States , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...