Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Am J Respir Crit Care Med ; 207(3): 261-270, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36099435

ABSTRACT

Rationale: There are limited therapeutic options for patients with coronavirus disease (COVID-19)-related acute respiratory distress syndrome with inflammation-mediated lung injury. Mesenchymal stromal cells offer promise as immunomodulatory agents. Objectives: Evaluation of efficacy and safety of allogeneic mesenchymal cells in mechanically-ventilated patients with moderate or severe COVID-19-induced respiratory failure. Methods: Patients were randomized to two infusions of 2 million cells/kg or sham infusions, in addition to the standard of care. We hypothesized that cell therapy would be superior to sham control for the primary endpoint of 30-day mortality. The key secondary endpoint was ventilator-free survival within 60 days, accounting for deaths and withdrawals in a ranked analysis. Measurements and Main Results: At the third interim analysis, the data and safety monitoring board recommended that the trial halt enrollment as the prespecified mortality reduction from 40% to 23% was unlikely to be achieved (n = 222 out of planned 300). Thirty-day mortality was 37.5% (42/112) in cell recipients versus 42.7% (47/110) in control patients (relative risk [RR], 0.88; 95% confidence interval, 0.64-1.21; P = 0.43). There were no significant differences in days alive off ventilation within 60 days (median rank, 117.3 [interquartile range, 60.0-169.5] in cell patients and 102.0 [interquartile range, 54.0-162.5] in control subjects; higher is better). Resolution or improvement of acute respiratory distress syndrome at 30 days was observed in 51/104 (49.0%) cell recipients and 46/106 (43.4%) control patients (odds ratio, 1.36; 95% confidence interval, 0.57-3.21). There were no infusion-related toxicities and overall serious adverse events over 30 days were similar. Conclusions: Mesenchymal cells, while safe, did not improve 30-day survival or 60-day ventilator-free days in patients with moderate and/or severe COVID-19-related acute respiratory distress syndrome.


Subject(s)
COVID-19 , Mesenchymal Stem Cells , Respiratory Distress Syndrome , Humans , COVID-19/therapy , SARS-CoV-2 , Lung , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/drug therapy
2.
J Control Release ; 337: 144-154, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34280414

ABSTRACT

One of the foremost challenges in translating nanoparticle technologies to the clinic is the requirement to produce materials on a large-scale. Scaling nanoparticle production methods is often non-trivial, and the success of these endeavors is frequently governed by whether or not an intermediate level of production, i.e., "pilot-scale" production, can be achieved. Pilot-scale production at the one-liter scale serves as a proof-of-concept that large-scale production will be possible. Here, we describe the pilot-scale production of the expansile nanoparticle (eNP) technology including verification of activity and efficacy following scaleup. We describe the challenges of sonication-based emulsification procedures and how these were overcome by use of a Microfluidizer technology. We also describe the problem-solving process that led to pre-polymerization of the nanoparticle polymer-a fundamental change from the lab-scale and previously published methods. Furthermore, we demonstrate good control over particle diameter, polydispersity and drug loading and the ability to sterilize the particles via filtration using this method. To facilitate long-term storage of these larger quantities of particles, we investigated six lyoprotectants and determined that sucrose is the most compatible with the current system. Lastly, we demonstrate that these changes to the manufacturing method do not adversely affect the swelling functionality of the particles, their highly specific localization to tumors, their non-toxicity in vivo or their efficacy in treating established intraperitoneal mesothelioma xenografts.


Subject(s)
Mesothelioma, Malignant , Mesothelioma , Nanoparticles , Humans , Polymers , Sonication
3.
J Cancer ; 10(15): 3344-3351, 2019.
Article in English | MEDLINE | ID: mdl-31293637

ABSTRACT

Inflammatory breast cancer (IBC) is an understudied and aggressive form of breast cancer with a poor prognosis, accounting for 2-6% of new breast cancer diagnoses but 10% of all breast cancer-related deaths in the United States. Currently there are no therapeutic regimens developed specifically for IBC, and it is critical to recognize that all aspects of treating IBC - including staging, diagnosis, and therapy - are vastly different than other breast cancers. In December 2014, under the umbrella of an interdisciplinary initiative supported by the Duke School of Medicine, researchers, clinicians, research administrators, and patient advocates formed the Duke Consortium for IBC to address the needs of patients in North Carolina (an ethnically and economically diverse state with 100 counties) and across the Southeastern United States. The primary goal of this group is to translate research into action and improve both awareness and patient care through collaborations with local, national and international IBC programs. The consortium held its inaugural meeting on Feb 28, 2018, which also marked Rare Disease Day and convened national research experts, clinicians, patients, advocates, government representatives, foundation leaders, staff, and trainees. The meeting focused on new developments and challenges in the clinical management of IBC, research challenges and opportunities, and an interactive session to garner input from patients, advocates, and community partners that would inform a strategic plan toward continuing improvements in IBC patient care, research, and education.

4.
Am J Transplant ; 19(7): 2122-2126, 2019 07.
Article in English | MEDLINE | ID: mdl-30913367

ABSTRACT

Abdominal wall transplantation (AWT) was introduced in 1999 in the context of reconstruction of complex abdominal wall defects in conjunction with visceral organ transplantation. As of recently, 38 cases of total AWT have been performed worldwide, about half of which were performed in the United States. While AWT is technically feasible, one of the major challenges presenting to the reconstructive surgeon is time to revascularization of the donor abdominal wall (AW), given the immediate proximity of the visceral organ and AWT. The authors report a novel AW revascularization technique during a synchronous small bowel and AWT in a 37-year-old man.


Subject(s)
Abdominal Wall/blood supply , Intestinal Fistula/therapy , Intestine, Small/transplantation , Organ Transplantation , Short Bowel Syndrome/therapy , Vascularized Composite Allotransplantation , Adult , Humans , Intestinal Fistula/pathology , Male , Prognosis , Short Bowel Syndrome/pathology
5.
Alzheimers Res Ther ; 3(5): 28, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21943025

ABSTRACT

Animal models have contributed significantly to our understanding of the underlying biological mechanisms of Alzheimer's disease (AD). As a result, over 300 interventions have been investigated and reported to mitigate pathological phenotypes or improve behavior in AD animal models or both. To date, however, very few of these findings have resulted in target validation in humans or successful translation to disease-modifying therapies. Challenges in translating preclinical studies to clinical trials include the inability of animal models to recapitulate the human disease, variations in breeding and colony maintenance, lack of standards in design, conduct and analysis of animal trials, and publication bias due to under-reporting of negative results in the scientific literature. The quality of animal model research on novel therapeutics can be improved by bringing the rigor of human clinical trials to animal studies. Research communities in several disease areas have developed recommendations for the conduct and reporting of preclinical studies in order to increase their validity, reproducibility, and predictive value. To address these issues in the AD community, the Alzheimer's Drug Discovery Foundation partnered with Charles River Discovery Services (Morrisville, NC, USA) and Cerebricon Ltd. (Kuopio, Finland) to convene an expert advisory panel of academic, industry, and government scientists to make recommendations on best practices for animal studies testing investigational AD therapies. The panel produced recommendations regarding the measurement, analysis, and reporting of relevant AD targets, th choice of animal model, quality control measures for breeding and colony maintenance, and preclinical animal study design. Major considerations to incorporate into preclinical study design include a priori hypotheses, pharmacokinetics-pharmacodynamics studies prior to proof-of-concept testing, biomarker measurements, sample size determination, and power analysis. The panel also recommended distinguishing between pilot 'exploratory' animal studies and more extensive 'therapeutic' studies to guide interpretation. Finally, the panel proposed infrastructure and resource development, such as the establishment of a public data repository in which both positive animal studies and negative ones could be reported. By promoting best practices, these recommendations can improve the methodological quality and predictive value of AD animal studies and make the translation to human clinical trials more efficient and reliable.

6.
Cancer Chemother Pharmacol ; 67(6): 1415-22, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20809121

ABSTRACT

PURPOSE: Corticorelin acetate (CrA) is a synthetic form of corticotropin-releasing factor undergoing clinical trials in the treatment of peritumoral brain edema (PBE). We sought to investigate preclinically its potential as an antitumor agent against human solid tumors and to assess its ability to enhance the therapeutic activity of bevacizumab (BEV) in these same models. METHODS: The in vivo efficacy of CrA as a single agent and in combination with the antiangiogenic agent, BEV, was examined in two preclinical human tumor models, the MX-1 breast and Colo-205 colon carcinomas. These models were selected based on their known sensitivity to BEV and were tumor types in which BEV has been approved for clinical use. The corneal micropocket assay was also performed to assess the antiangiogenic activity of CrA relative to BEV. The exposure level of CrA in the mouse using a typical preclinical regimen was measured so as to compare it to reported clinical exposure levels. RESULTS: CrA was active as a single agent in the MX-1 breast carcinoma, but did not exhibit statistically significant activity as a single agent in the Colo-205 colon carcinoma under the doses and schedules used in the study. When BEV, which was active or near active in both the MX-1 and Colo-205 models, was administered concomitantly with CrA, therapeutic outcomes were observed that were significantly better than those obtained using either monotherapy. These therapeutic potentiations using CrA plus BEV were obtained in the absence of any observable increase in toxicities. CrA was active in the corneal micropocket assay, producing a substantial (>70%) inhibition of neovascularization. A representative CrA regimen in mice produced an exposure within eightfold of human exposure determined at one-half the current clinical dose. CONCLUSIONS: The application of CrA for the treatment of PBE likely involves its activity as an antiangiogenic agent, which may be one possible mechanism to explain its observed preclinical antitumor activity. That activity, as well as its ability to provide an enhanced therapeutic outcome when given in conjunction with BEV in the absence of increased toxicity, supports the use of CrA clinically as other than a replacement therapy for dexamethasone in PBE.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Colonic Neoplasms/drug therapy , Corticotropin-Releasing Hormone/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab , Corticotropin-Releasing Hormone/administration & dosage , Corticotropin-Releasing Hormone/pharmacokinetics , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous
7.
J Med Chem ; 47(22): 5367-80, 2004 Oct 21.
Article in English | MEDLINE | ID: mdl-15481975

ABSTRACT

Two closely related diaryl acylsulfonamides were recently reported as potent antitumor agents against a broad spectrum of human tumor xenografts (colon, lung, breast, ovary, and prostate) in nude mice. Especially intriguing was their activity against colorectal cancer xenografts. In this paper, rapid parallel synthesis along with traditional medicinal chemistry techniques were used to quickly delineate the structure-activity relationships of the substitution patterns in both phenyl rings of the acylsufonamide anti-proliferative scaffold. Although the molecular target of the compounds remains unclear, we determined that the vascular endothelial growth factor-dependent human umbilical vein endothelial cells assay in combination with a soft agar disk diffusion assay allowed for optimization of potency in the series. The pharmacokinetic properties and in vivo activity in an HCT116 xenograft model are reported for representative compounds.


Subject(s)
Antineoplastic Agents/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Line , Drug Screening Assays, Antitumor , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Female , Half-Life , Humans , In Vitro Techniques , Mice , Mice, Nude , Quantitative Structure-Activity Relationship , Rats , Rats, Inbred F344 , Sulfonamides/chemistry , Sulfonamides/pharmacology , Transplantation, Heterologous , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/physiology
8.
Int J Oncol ; 21(1): 65-72, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12063551

ABSTRACT

GEM 231, a second-generation antisense oligonucleotide targeted against the RIalpha subunit of protein kinase A (PKA) was co-administered with the chemotherapeutic agent irinotecan, a topoisomerase-I inhibitor, to study the antitumor efficacy of the combination in nude mice bearing various human tumor xenografts. The combination treatment of GEM 231 and irinotecan produced enhanced and prolonged tumor-growth inhibition, compared with irinotecan monotherapy, against human colon (HCT-116), pancreas (Panc-1), prostate (PC3) and lung (SKMES) tumors in mice. The extent of tumor-growth inhibition, however, varied among the different tumor models studied. The tumor-growth inhibition depended on the dose of GEM 231 co-administered with irinotecan. The combination of GEM 231 (20 mg/kg, i.p., 5 days on 2 days off x 7) and irinotecan (50 mg/kg, i.v., qwk x 3) produced significantly longer tumor-growth delay than did irinotecan administered alone. Importantly, the co-administration of irinotecan and GEM 231 did not result in higher toxicity compared with monotherapies in the several tumor models tested. These results suggest that the use of irinotecan in combination with GEM 231 may increase the therapeutic index of irinotecan in cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Oligonucleotides, Antisense/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Body Weight/drug effects , Camptothecin/administration & dosage , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Dose-Response Relationship, Drug , Female , Humans , Irinotecan , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Oligonucleotides/administration & dosage , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Survival Rate , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...