Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Rheumatology (Oxford) ; 55(11): 1993-2000, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27498355

ABSTRACT

OBJECTIVES: Binding immunoglobulin protein (BiP) is a human endoplasmic reticulum-resident stress protein. In pre-clinical studies it has anti-inflammatory properties due to the induction of regulatory cells. This randomized placebo-controlled, dose ascending double blind phase I/IIA trial of BiP in patients with active RA, who had failed accepted therapies, had the primary objective of safety. Potential efficacy was measured by DAS28-ESR and changes in biomarkers. METHODS: Twenty-four patients with active RA who had failed one or more DMARDs were sequentially assigned to three groups each of eight patients randomly allocated to receive placebo (two patients) or BiP (six patients), 1, 5 or 15 mg. Patients received a single i.v. infusion over 1 h and were observed as inpatients overnight. A 12-week follow-up for clinical, rheumatological and laboratory assessments for safety, efficacy (DAS28-ESR) and biomarker analysis was performed. RESULTS: No infusion reactions or serious adverse drug reactions were noted. Adverse events were evenly distributed between placebo and BiP groups with no BiP-related toxicities. Haematological, renal and metabolic parameters showed no drug-related toxicities. Remission was only achieved by patients in the 5 and 15 mg groups, and not patients who received placebo or 1 mg BiP. Good DAS28-ESR responses were achieved in all treatment groups. The BiP responding patients showed significantly lower serum concentrations of CRP, 2 weeks post-infusion compared with pre-infusion levels, and of VEGF and IL-8 from the placebo group. CONCLUSION: BiP (⩽15 mg) is safe in patients with active RA. Some patients had clinical and biological improvements in RA activity. BiP merits further study. TRIAL REGISTRATION: ISRCTN registry, http://isrctn.com, ISRCTN22288225 and EudraCT, https://eudract.ema.europa.eu, 2011-005831-19.


Subject(s)
Antirheumatic Agents/administration & dosage , Arthritis, Rheumatoid/drug therapy , Lymphokines/administration & dosage , Adolescent , Adult , Aged , Antirheumatic Agents/adverse effects , Biological Products/therapeutic use , Biomarkers/metabolism , Double-Blind Method , Female , Humans , Infusions, Intravenous , Interleukin-8/metabolism , Lymphokines/adverse effects , Male , Middle Aged , Recombinant Proteins , Remission Induction , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism , Young Adult
2.
Rev. AMRIGS ; 54(1): 72-76, jan.-mar. 2010. ilus
Article in Portuguese | LILACS | ID: lil-685588

ABSTRACT

O presente relato de caso tem como finalidade chamar a atenção de doença grave que frequentemente é confundida com septicemia, no entanto o mecanismo etiológico é decorrente de defeitos genéticos ou associados à resposta imunológica exagerada, decorrente de ação citotóxica de linfócitos T CD8 e histiócitos, acarretando proliferação clonal e ativação de células ”natural killer” (NK). Uma tempestade de linfocinas acontece e como consequência é iniciada uma incontrolável hemofagocitose de todos os elementos sanguíneos, terminando pela infecção secundária do organismo por ausência de destruição de patógenos. A maioria dos casos termina pela morte do paciente; no entanto, relatamos nesse caso a possibilidade de incluirmos a plasmaferese como forma de retirar as linfocinas circulantes, razão do estímulo à destruição celular. O tratamento concomitante com alta dose de imunoglobulina endovenosa também foi realizado


The purpose of the present case report is to call attention to a serious disease that is often mistaken with septicemia, although its etiological mechanism results from genetic defects or is associated with an immune over-reaction, resulting from cytotoxic action of CD8 T lymphocytes and histiocytes, causing clonal proliferation and activation of “natural killer” (NK) cells. There occurs a storm of lymphokines and, as a consequence, an uncontrollable hemophagocytosis of all blood elements, which leads to secondary infection of the organism because of absence of pathogens destruction. Although most of the cases end up in death, in this case we report the possibility of including plasmapheresis as a way to remove the circulating lymphokines, the reason for stimulation of cell destruction. Co-treatment with high dose of intravenous immunoglobulin was performed too


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Immunoglobulins, Intravenous/therapeutic use , Lymphokines/adverse effects , Lymphokines/poisoning , Plasmapheresis
3.
Circulation ; 108(16): 1933-8, 2003 Oct 21.
Article in English | MEDLINE | ID: mdl-14504183

ABSTRACT

BACKGROUND: "Therapeutic angiogenesis" seeks to improve perfusion by the growth of new blood vessels. The Regional Angiogenesis with Vascular Endothelial growth factor (RAVE) trial is the first major randomized study of adenoviral vascular endothelial growth factor (VEGF) gene transfer for the treatment of peripheral artery disease (PAD). METHODS AND RESULTS: This phase 2, double-blind, placebo-controlled study was designed to test the efficacy and safety of intramuscular delivery of AdVEGF121, a replication-deficient adenovirus encoding the 121-amino-acid isoform of vascular endothelial growth factor, to the lower extremities of subjects with unilateral PAD. In all, 105 subjects with unilateral exercise-limiting intermittent claudication during 2 qualifying treadmill tests, with peak walking time (PWT) between 1 to 10 minutes, were stratified on the basis of diabetic status and randomized to low-dose (4x10(9) PU) AdVEGF121, high-dose (4x10(10) PU) AdVEGF121, or placebo, administered as 20 intramuscular injections to the index leg in a single session. The primary efficacy end point, change in PWT (DeltaPWT) at 12 weeks, did not differ between the placebo (1.8+/-3.2 minutes), low-dose (1.6+/-1.9 minutes), and high-dose (1.5+/-3.1 minutes) groups. Secondary measures, including DeltaPWT, ankle-brachial index, claudication onset time, and quality-of-life measures (SF-36 and Walking Impairment Questionnaire), were also similar among groups at 12 and 26 weeks. AdVEGF121 administration was associated with increased peripheral edema. CONCLUSIONS: A single unilateral intramuscular administration of AdVEGF121 was not associated with improved exercise performance or quality of life in this study. This study does not support local delivery of single-dose VEGF121 as a treatment strategy in patients with unilateral PAD.


Subject(s)
Adenoviridae/genetics , Endothelial Growth Factors/administration & dosage , Genetic Vectors/administration & dosage , Intercellular Signaling Peptides and Proteins/administration & dosage , Lymphokines/administration & dosage , Neovascularization, Physiologic/drug effects , Peripheral Vascular Diseases/therapy , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Edema/chemically induced , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/genetics , Female , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Intercellular Signaling Peptides and Proteins/adverse effects , Intercellular Signaling Peptides and Proteins/genetics , Intermittent Claudication/etiology , Intermittent Claudication/therapy , Lymphokines/adverse effects , Lymphokines/genetics , Male , Middle Aged , Peripheral Vascular Diseases/complications , Quality of Life , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Walking/statistics & numerical data
4.
Circulation ; 107(10): 1359-65, 2003 Mar 18.
Article in English | MEDLINE | ID: mdl-12642354

ABSTRACT

BACKGROUND: Recombinant human vascular endothelial growth factor protein (rhVEGF) stimulates angiogenesis in animal models and was well tolerated in Phase I clinical trials. VIVA (Vascular endothelial growth factor in Ischemia for Vascular Angiogenesis) is a double-blind, placebo-controlled trial designed to evaluate the safety and efficacy of intracoronary and intravenous infusions of rhVEGF. METHODS AND RESULTS: A total of 178 patients with stable exertional angina, unsuitable for standard revascularization, were randomized to receive placebo, low-dose rhVEGF (17 ng x kg(-1) x min(-1)), or high-dose rhVEGF (50 ng x kg(-1) x min(-1)) by intracoronary infusion on day 0, followed by intravenous infusions on days 3, 6, and 9. Exercise treadmill tests, angina class, and quality of life assessments were performed at baseline, day 60, and day 120. Myocardial perfusion imaging was performed at baseline and day 60. At day 60, the change in exercise treadmill test (ETT) time from baseline was not different between groups (placebo, +48 seconds; low dose, +30 seconds; high dose, +30 seconds). Angina class and quality of life were significantly improved within each group, with no difference between groups. By day 120, placebo-treated patients demonstrated reduced benefit in all three measures, with no significant difference compared with low-dose rhVEGF. In contrast, high-dose rhVEGF resulted in significant improvement in angina class (P=0.05) and nonsignificant trends in ETT time (P=0.15) and angina frequency (P=0.09) as compared with placebo. CONCLUSIONS: rhVEGF seems to be safe and well tolerated. rhVEGF offered no improvement beyond placebo in all measurements by day 60. By day 120, high-dose rhVEGF resulted in significant improvement in angina and favorable trends in ETT time and angina frequency.


Subject(s)
Endothelial Growth Factors/therapeutic use , Intercellular Signaling Peptides and Proteins/therapeutic use , Lymphokines/therapeutic use , Myocardial Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Adult , Aged , Angina Pectoris/classification , Coronary Circulation , Coronary Vessels/drug effects , Coronary Vessels/growth & development , Double-Blind Method , Endothelial Growth Factors/administration & dosage , Endothelial Growth Factors/adverse effects , Exercise Test , Female , Humans , Infusions, Intravenous , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/adverse effects , Lymphokines/administration & dosage , Lymphokines/adverse effects , Male , Middle Aged , Myocardial Ischemia/diagnosis , Quality of Life , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
Blood ; 102(1): 161-8, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12649136

ABSTRACT

Gene therapy approaches involving vascular endothelial growth factor (VEGF) to promote therapeutic angiogenesis are under consideration for conditions ranging from ischemic heart disease to nonhealing skin ulcers. Here we make the surprising observation that the transgenic delivery of VEGF to the skin results in a profound inflammatory skin condition with many of the cellular and molecular features of psoriasis, including the characteristic vascular changes, epidermal alterations, and inflammatory infiltrates. Even longstanding psoriatic disease remains dependent on the transgenic VEGF in this model because it can be effectively reversed by the addition of VEGF Trap, a potent VEGF antagonist. Previous attempts to faithfully replicate the psoriatic phenotype through the transgenic delivery of epidermal keratinocyte growth factors or inflammatory mediators generated phenotypes with only partial resemblance to human psoriasis, leaving unanswered questions about the etiology of this disease. The ability of transgenic VEGF to induce a psoriasiform phenotype suggests a new etiology and treatment approach for this disease and further substantiates emerging concerns about possible proinflammatory adverse effects that might be associated with therapeutic attempts to deliver VEGF.


Subject(s)
Endothelial Growth Factors/administration & dosage , Endothelial Growth Factors/adverse effects , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/adverse effects , Lymphokines/administration & dosage , Lymphokines/adverse effects , Psoriasis/chemically induced , Animals , Blood Vessels/drug effects , Blood Vessels/pathology , Disease Models, Animal , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Immunohistochemistry , Inflammation/chemically induced , Inflammation/etiology , Mice , Mice, Transgenic , Psoriasis/etiology , Receptors, Growth Factor , Recombinant Fusion Proteins/pharmacology , Skin/drug effects , Skin/injuries , Skin/pathology , Transgenes , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
6.
Am J Cardiol ; 90(5): 512-6, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12208412

ABSTRACT

The long-term safety and efficacy of adenoviral delivery of growth factors in patients with peripheral arterial disease (PAD) is unknown. CI-1023 (Ad(GV)VEGF(121.10)) is a replication-deficient adenovirus encoding human vascular endothelial growth factor isoform 121. In this phase I trial, we investigated the safety and efficacy of CI-1023 in subjects with advanced claudication symptoms secondary to infra-inguinal disease. Eighteen subjects >35 years of age with a median ankle brachial index (ABI) at rest of 0.525 (interquartile range 0.4) and angiographic disease involving the infra-inguinal vessels underwent intramuscular injection of CI-1023 (4 x10(8) to 4 x10(10) particle units, n = 15) or placebo (n = 3). Eleven of 15 patients (73%) who received CI-1023 and 1 of 3 subjects (33%) who received placebo, completed 1 year of follow-up. Edema and rash were the most common early adverse event. One infra-inguinal bypass procedure occurred in each of the placebo and CI-1023 groups at days 29 and 104, respectively. One death (day 160) and 1 malignancy (day 274) occurred in the CI-1023 group. Conclusions on efficacy could not be made due to the small number of patients. However, there were encouraging trends in ABI at rest and peak walking time at follow-up.


Subject(s)
Adenoviridae/physiology , DNA, Complementary/administration & dosage , Endothelial Growth Factors/administration & dosage , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , Intermittent Claudication/drug therapy , Lymphokines/administration & dosage , Virus Replication/physiology , Adult , Aged , DNA, Complementary/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/blood , Extremities/blood supply , Extremities/pathology , Female , Follow-Up Studies , Genetic Vectors/adverse effects , Humans , Immune System/drug effects , Immune System/immunology , Intermittent Claudication/epidemiology , Lymphokines/adverse effects , Lymphokines/blood , Male , Middle Aged , Risk Factors , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Walking
7.
Hypertension ; 39(6): 1095-100, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12052848

ABSTRACT

Vascular endothelial growth factor (VEGF) exerts vasodilation-induced hypotension as a major side effect for treatment of ischemic diseases. VEGF has 2 receptor tyrosine kinases, KDR and Flt-1. Little is known about which receptor mediates VEGF-induced hypotension. To elucidate the role of each receptor in mediating hypotension, KDR-selective and Flt-1-selective mutants were used for in vitro and in vivo studies. The KDR-selective mutant induced vascular endothelial cell proliferation comparable to VEGF, whereas the Flt-1- selective mutant had no effect on proliferation. Intravenous injection of KDR-selective mutant, Flt-selective mutant, or VEGF caused a dose-related decrease in mean arterial pressure in conscious rats. The hypotensive response to KDR-selective mutant was significantly less than that to VEGF (P<0.01) but was greater than that to Flt-selective mutant (P<0.01). Similarly, VEGF and KDR-selective mutant induced more potent vasorelaxation than Flt-selective mutant or placenta growth factor that binds Flt-1 only (P<0.01), and the vasorelaxation to KDR-selective mutant was not significantly different at low concentrations but less than that to VEGF at high concentrations. The results indicate that the vasodilation and hypotensive effect of VEGF may involve both receptors, but KDR is the predominant receptor mediating this effect. Because KDR-selective mutant induced proliferation and angiogenesis similar to VEGF but was associated with 36% attenuation in hypotension, the data suggest that the KDR-selective mutant may represent an alternative treatment for ischemic diseases.


Subject(s)
Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/pharmacology , Hypotension/etiology , Lymphokines/adverse effects , Lymphokines/pharmacology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/physiology , Vasodilator Agents/adverse effects , Vasodilator Agents/pharmacology , Animals , Aorta/physiopathology , Blood Pressure/drug effects , Cell Division/drug effects , Cells, Cultured , Culture Techniques , Dose-Response Relationship, Drug , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Humans , Hypotension/physiopathology , Lymphokines/genetics , Lymphokines/metabolism , Male , Mutation , Proto-Oncogene Proteins/physiology , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors , Vasodilation/drug effects , Vasodilator Agents/metabolism
8.
Neurosurgery ; 50(3): 589-98, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11841728

ABSTRACT

OBJECTIVE: Therapeutic cerebral angiogenesis, i.e., using angiogenic factors to enhance collateral vessel formation within the central nervous system, is a potential method for cerebral revascularization. Vascular endothelial growth factor (VEGF) is a potent endothelial cell mitogen that also increases capillary permeability, particularly in ischemic tissue. The purpose of this study was to assess the angiogenic and capillary permeability effects of chronic intraventricular infusion of exogenous VEGF in nonischemic brain tissue, because many patients with impaired cerebrovascular reserve do not exhibit chronic cerebral ischemia. METHODS: Recombinant human VEGF(165) was infused into the right lateral ventricle of rats at a rate of 1 microl/h for 7 days, at concentrations of 1 to 25 microg/ml, with osmotic minipumps. Control animals received vehicle only. Vessels were identified in laminin immunohistochemical analyses. Capillary permeability and brain edema were assessed with Evans blue extravasation, [(3)H]inulin permeability, and brain water content measurements. RESULTS: Vessel density was dose-dependently increased by VEGF(165) infusions, with significant increases occurring in animals treated with 5 or 25 microg/ml, compared with control animals (P h 0.01). Significant enlargement of the lateral ventricles was observed for the highest-dose group but not for animals treated with other doses. Capillary permeability was assessed in animals treated with a dose of 5 microg/ml. An increase in capillary permeability in the diencephalon was identified with Evans blue extravasation and [(3)H]inulin permeability assessments; however, the brain water content was not significantly increased. CONCLUSION: Chronic intraventricular infusions of VEGF(165) increased vascular density in a dose-dependent manner. There seems to be a therapeutic window, because infusion of VEGF(165) at a concentration of 5 microg/ml resulted in a significant increase in vessel density with minimal associated brain edema and no ventriculomegaly.


Subject(s)
Cerebrovascular Circulation/drug effects , Endothelial Growth Factors/administration & dosage , Lymphokines/administration & dosage , Neovascularization, Physiologic/drug effects , Animals , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Body Water/metabolism , Brain/drug effects , Brain/pathology , Brain Edema/chemically induced , Coloring Agents/pharmacokinetics , Dose-Response Relationship, Drug , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/pharmacology , Evans Blue/pharmacokinetics , Humans , Injections, Intraventricular , Inulin/pharmacokinetics , Lymphokines/adverse effects , Lymphokines/pharmacology , Permeability , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Rev Esp Cardiol ; 54(10): 1210-24, 2001 Oct.
Article in Spanish | MEDLINE | ID: mdl-11591302

ABSTRACT

Therapeutic angiogenesis based on the administration of growth factors with angiogenic activity allows enhancement of collateral vessels able to palliate insufficient tissue perfusion secondary to obstruction of native arteries. At present, this type of therapy is addressed to patients that fail to respond to conventional treatment (surgical or percutaneous revascularization). The most extensively investigated angiogenic growth factors are vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF). These cytokines can be administered either as recombinant proteins or as the genes encoding for these proteins. Both approaches have pros and cons that are under investigation in animal models and in clinical studies. Although clinical trials consist so far of small, often non-randomized series, preliminary results are promising. For example, administration of VEGF or FGF has been associated to objective evidence of increased tissue perfusion in patients with myocardial ischemia, and to a significant improvement of pain and ischemia in patients with peripheral arterial disease. Contrarily to expected, these interventions have been associated to scant adverse side effects, although larger clinical trials will be necessary in order to prove the safety and effectiveness of these interventions. Nevertheless, it seems clear that it is feasible to induce effective therapeutic angiogenesis in selected patients without significant associated toxicity.


Subject(s)
Endothelial Growth Factors/therapeutic use , Fibroblast Growth Factors/therapeutic use , Ischemia/therapy , Lymphokines/therapeutic use , Peripheral Vascular Diseases/therapy , Clinical Trials as Topic , Drug Evaluation , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/genetics , Fibroblast Growth Factors/adverse effects , Fibroblast Growth Factors/genetics , Genetic Therapy/methods , Humans , Ischemia/drug therapy , Leg/blood supply , Lymphokines/adverse effects , Lymphokines/genetics , Myocardial Ischemia/drug therapy , Neovascularization, Physiologic , Secondary Prevention , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Curr Opin Investig Drugs ; 2(6): 792-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11572658

ABSTRACT

GenVec, in collaboration with Pfizer (formerly Parke-Davis), is developing AdGVVEGF121.10 (BioBypass), a gene therapy involving the 121-residue isoform of vascular endothelial growth factor (VEGF), licensed from Scios, for the potential treatment of coronary artery disease (CAD) and peripheral vascular disease (PVD) [262000]. By March 2000, phase II trials in CAD had commenced [359531], [359532], [359538]. By August 2000, phase II trials were also underway for PVD [386293]. The initial phase II trial will include approximately 70 patients with severe CAD who are not candidates for bypass surgery and will assess exercise capacity and patient well-being, before and after treatment, as well as safety and drug tolerance [364137]. Scios licensed the gene transfer applications of VEGF121 to GenVec in June 1996 [263381]. In September 1997, GenVec entered into an agreement with Parke-Davis, a subsidiary of Warner-Lambert (now Pfizer), to develop the therapy [262000]. In May 1999, Warner-Lambert signed an agreement with Bioscience for a device for the administration of AdGVVEGF121.10 1325443]. In May 2000, Merrill Lynch predicted a US filing in the first half of 2003 [375962]. In January 2001, AG Edwards predicted the product would generate $70 million in revenues to Pfizer and $12 million in royalties to GenVec in 2005. In February 1999, GenVec was awarded US-05846782, covering vectors for targeting the transfer of therapeutic genes to specific tissues in the human body [316038].


Subject(s)
Angiogenic Proteins , Endothelial Growth Factors/pharmacology , Genetic Therapy , Lymphokines/pharmacology , Vascular Diseases/therapy , Animals , Clinical Trials as Topic , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/pharmacokinetics , Humans , Lymphokines/adverse effects , Lymphokines/pharmacokinetics , Structure-Activity Relationship
12.
Bone Marrow Transplant ; 27(11): 1173-80, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11551028

ABSTRACT

Hepatic veno-occlusive disease (VOD) is one of the most serious complications in patients receiving stem cell transplantation (SCT). However, the cause of VOD remained to be elucidated. Vascular endothelial growth factor (VEGF) has been reported to have various physiological effects including neovascularization and acceleration of vasopermeability. Because we postulated that VEGF could be one of the causative factors in VOD after SCT, serum VEGF levels were measured by ELISA in 50 patients receiving SCT. Six of the patients showed typical manifestations of VOD and four of them died due to VOD. The mean maximum serum VEGF level in the six patients with VOD was markedly increased compared to that in the patients without VOD (P < 0.001) and in normal controls (P < 0.001). Moreover, the mean maximum serum VEGF level in patients with VOD before conditioning chemoradiotherapy for SCT was also high compared to patients without VOD (P = 0.0012) in the same period. Similarly, serum VEGF levels were significantly higher in patients whose plasma protein C activities decreased below 40% (P < 0.001). During the clinical course of VOD after SCT, the increase of serum VEGF synchronized fairly well with the development of VOD. Since VEGF causes the expression of tissue factor on circulating monocyte/macrophages and results in hypercoagulability, our observation suggests that in the patients with VOD who showed high serum VEGF it might account for the development of VOD. Furthermore, this observation may indicate a novel therapeutic strategy for prevention of VOD.


Subject(s)
Endothelial Growth Factors/blood , Hematopoietic Stem Cell Transplantation/adverse effects , Hepatic Veno-Occlusive Disease/blood , Hepatic Veno-Occlusive Disease/etiology , Lymphokines/blood , Adolescent , Adult , Biomarkers/blood , Child , Child, Preschool , Combined Modality Therapy , Cytokines/adverse effects , Cytokines/blood , Endothelial Growth Factors/adverse effects , Enzyme-Linked Immunosorbent Assay , Female , Hematologic Diseases/complications , Hematologic Diseases/therapy , Hepatic Veno-Occlusive Disease/diagnosis , Humans , Infant , Lymphokines/adverse effects , Male , Prognosis , Protein C/metabolism , Time Factors , Transplantation Conditioning , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
J Am Coll Cardiol ; 37(8): 2126-30, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11419898

ABSTRACT

OBJECTIVES: This study was designed to evaluate the impact of recombinant human vascular endothelial growth factor165 (rhVEGF) on atherosclerotic plaque progression. BACKGROUND: Therapeutic angiogenesis represents a promising treatment for ischemic diseases. However, angiogenesis may impact atherosclerosis. METHODS: Albumin or rhVEGF was administered by a single intramuscular injection (2 microg/kg body weight) to New Zealand White rabbits fed with a 0.25% cholesterol diet beginning three weeks before therapy. Subsets of rabbits from each group underwent perfusion-fixation and harvesting of the thoracic aorta for morphometric and immunohistochemical analyses at 7 or 21 days. RESULTS: The mean plaque area was 15.75+/-2.28% and 22.00+/-3.24% with VEGF and 0.67+/-0.22% and 1.17+/-0.34% with albumin at 7 and 21 days, respectively. The plaque circumference was 13.00+/-2.58% and 23.75+/-2.86% with VEGF and 2.50+/-0.65% and 6.25+/-1.88% with albumin at 7 and 21 days, respectively. The maximal plaque thickness was 0.11+/-0.002 and 0.15+/-0.007 mm with VEGF and 0.04+/-0.009 and 0.07+/-0.003 mm with albumin at 7 and 21 days, respectively. The endothelial density (reported as percent total plaque area) was 31.75+/-4.42% and 63.00+/-8.45% with VEGF and 7.75+/-1.65% and 12.75+/-1.93% with albumin at 7 and 21 days, respectively. The macrophage density was 4.5+/-0.86 and 19.25+/-1.54 with VEGF and 4.26+/-0.75 and 6.00+/-1.08 with albumin at 7 and 21 days, respectively. CONCLUSIONS: Recombinant human VEGF increases the rate and degree of atherosclerotic plaque formation in the thoracic aorta in a cholesterol-fed rabbit model.


Subject(s)
Aortic Diseases/pathology , Arteriosclerosis/pathology , Endothelial Growth Factors/adverse effects , Lymphokines/adverse effects , Protein Isoforms/adverse effects , Animals , Aorta, Thoracic , Arteriosclerosis/physiopathology , Disease Progression , Immunohistochemistry , Macrophages , Neovascularization, Physiologic , Rabbits , Recombinant Proteins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
17.
Leuk Lymphoma ; 41(3-4): 229-37, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11378536

ABSTRACT

Primary effusion lymphomas (PEL), rare lymphomas associated with Kaposi's sarcoma-associated herpesvirus (KSHV or HHV-8) infection, present as malignant lymphomatous effusions in body cavities. We have recently found that PEL effusions contain high levels of vascular endothelial growth factor/vascular permeability factor (VEGF/VPF). VEGF/VPF, an important regulator of tumor-angiogenesis in vivo, exerts its effects acting through the receptors KDR/Flk-1 and Flt-1 on the endothelial cell membrane. In vitro, the PEL cell lines BC-1, BCP-1 and BCBL-1 produce high levels of VEGF. RT-PCR analysis of RNA from the PEL cell lines amplified the three VEGF/VPF secreted isoforms, VEGF/VPF(121), VEGF/VPF(145) and VEGF/VPF(165). Two of the PEL cell lines express the VEGF/VPF receptor Flt-1, but VEGF did not stimulate proliferation in these cells. SCID/beige mice inoculated intraperitoneally with BCBL-1 cells developed effusion lymphoma of human cell origin with prominent bloody ascites. In contrast, none of the mice treated with a neutralizing anti-human VEGF/VPF antibody developed ascites and effusion lymphoma. Although the precise mechanisms by which VEGF/VPF can promote vascular permeability are not fully understood, VEGF/VPF stimulation of vascular leakage may be critical to the pathogenesis of PEL.


Subject(s)
Endothelial Growth Factors/physiology , Lymphokines/physiology , Lymphoma, AIDS-Related/etiology , Animals , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/metabolism , Herpesviridae Infections/complications , Herpesvirus 8, Human , Humans , Lymphokines/adverse effects , Lymphokines/metabolism , Lymphoma, AIDS-Related/metabolism , Peritoneal Neoplasms/etiology , Peritoneal Neoplasms/metabolism , Pleural Effusion, Malignant/etiology , Pleural Effusion, Malignant/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
19.
Cardiovasc Res ; 49(3): 532-42, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11166266

ABSTRACT

The importance of spontaneously developing collateral vessels to supplement perfusion of tissue rendered ischemic by vascular obstruction was recognized many years ago. However, it was not until potent angiogenesis factors were identified, purified, and produced in sufficient quantities, that the field began its rapid development. In the early 1990s it was first shown that basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) proteins could actually stimulate collateral flow. However, additional studies also demonstrated that the duration of exposure of the vessels to angiogenesis factors was critical, and that the administration of proteins, with their relatively brief half-lives, may pose important practical limitations. The demonstration that gene therapy can improve collateral function presents one of the solutions to the conundrum, since gene therapy can be considered a sophisticated form of a sustained delivery system. The results of several clinical trials have been reported. All involve administration of single angiogenesis agents, and most are Phase I trials. The two studies rising to Phase II status demonstrated no treatment effect on the primary end-point. It may therefore be relevant to consider that the molecular mechanisms responsible for angiogenesis are extraordinarily complex, and an optimal angiogenesis intervention may require a 'multiple factor' strategy. It is important to note that no serious side-effects ascribable to an angiogenesis agent were recognized in these trials. However, angiogenesis agents are potent molecules with multiple activities. It is therefore possible that they might occasionally cause side-effects, some serious. Among these, based on their biologic activities, are neovascularization of non-targeted tissues, expansion and induction of instability of atherogenic plaque, and growth of tumors. In summary, there is ample experimental evidence justifying an optimistic outlook relating to our eventually being successful in enhancing collateral flow to ischemic tissue in a clinical setting. However, we are not there yet, and identification of the optimal angiogenesis strategy is still unclear. Additional experimental work, in parallel with large, carefully controlled clinical trials are needed to continue the exciting advances of the last decade, and to achieve the goal of providing patients with alternative potent therapies to improve collateral flow, and thereby to alleviate their symptoms and perhaps to prolong their lives.


Subject(s)
Collateral Circulation , Coronary Circulation , Coronary Disease/therapy , Growth Substances/therapeutic use , Neovascularization, Physiologic , Animals , Capillary Permeability/drug effects , Disease Models, Animal , Dogs , Endothelial Growth Factors/adverse effects , Endothelial Growth Factors/genetics , Endothelial Growth Factors/therapeutic use , Fibroblast Growth Factor 2/adverse effects , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/therapeutic use , Gene Expression , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/adverse effects , Growth Substances/adverse effects , Growth Substances/genetics , Hypotension/chemically induced , Lymphokines/adverse effects , Lymphokines/genetics , Lymphokines/therapeutic use , Randomized Controlled Trials as Topic , Stress, Mechanical , Transgenes , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...