Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Biomed Pharmacother ; 110: 803-817, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30554119

ABSTRACT

Cancer stem cells (CSCs) are the promising targets for cancer chemotherapy that cannot be eliminated by conventional chemotherapy. In this study cationic liposomes of cabazitaxel (CBX) and silibinin (SIL) were prepared with an aim to kill cancer cells and CSCs for prostate cancer. CBX act as cancer cell inhibitor and SIL as CSC inhibitor. Hyaluronic acid (HA), an endogenous anionic polysaccharide was coated on cationic liposomes for targeting CD44 receptors over expressed on CSCs. Liposomes were prepared by ethanol injection method with particle size below 100 nm and entrapment efficiency of more than 90% at 10% w/w drug loading. Liposomes were characterized by dynamic light scattering, transmission electron microscopy, 1H nuclear magnetic resonance and scanning electron microscopy-energy dispersive x-ray spectroscopy. Liposomes were evaluated for their anticancer action in androgen independent human prostate cancer cell lines (PC-3 and DU-145). HA coated liposomes showed potential cytotoxicity over other groups with low IC50, significantly inhibited cell migration and induced apoptosis. Synergistic cytotoxic effect was also observed with HA coated liposomes that resulted in colony formation inhibition and G2/M phase arrest. Proficient cytotoxicity against CD44+ cells (14.87 ± 0.41% in PC-3 and 33.95 ± 0.68% in DU-145 cells) indicated the efficiency of HA coated liposomes towards CSC targeting. Hence, the outcome of this combinational therapy with CD44 targeting indicates the suitability of HA coated CBX and SIL co-loaded liposomes as a potential approach for eradicating prostate cancer and herein might provide a insight for future studies.


Subject(s)
Drug Delivery Systems/methods , Hyaluronan Receptors/administration & dosage , Nanomedicine/methods , Prostatic Neoplasms , Silybin/administration & dosage , Taxoids/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cations , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hyaluronan Receptors/metabolism , Liposomes , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Silybin/pharmacokinetics , Taxoids/pharmacokinetics , Tumor Stem Cell Assay/methods
2.
Tumour Biol ; 39(5): 1010428317701628, 2017 May.
Article in English | MEDLINE | ID: mdl-28468593

ABSTRACT

Peritoneal carcinomatosis results from dissemination of solid tumors in the peritoneal cavity, and is a common site of metastasis in patients with carcinomas of gastrointestinal or gynecological origin. Peritoneal carcinomatosis treatment is challenging as poorly vascularized, disseminated peritoneal micro-tumors are shielded from systemic anticancer drugs and drive tumor regrowth. Here, we describe the identification and validation of a tumor homing peptide CKRDLSRRC (IP3), which upon intraperitoneal administration delivers payloads to peritoneal metastases. IP3 peptide was identified by in vivo phage display on a mouse model of peritoneal carcinomatosis of gastric origin (MKN-45P), using high-throughput sequencing of the peptide-encoding region of phage genome as a readout. The IP3 peptide contains a hyaluronan-binding motif, and fluorescein-labeled IP3 peptide bound to immobilized hyaluronan in vitro. After intraperitoneal administration in mice bearing peritoneal metastases of gastric and colon origin, IP3 peptide homed robustly to macrophage-rich regions in peritoneal tumors, including poorly vascularized micro-tumors. Finally, we show that IP3 functionalization conferred silver nanoparticles the ability to home to peritoneal tumors of gastric and colonic origin, suggesting that it could facilitate targeted delivery of nanoscale payloads to peritoneal tumors. Collectively, our study suggests that the IP3 peptide has potential applications for targeting drugs, nanoparticles, and imaging agents to peritoneal tumors.


Subject(s)
Carcinoma/drug therapy , Hyaluronan Receptors/administration & dosage , Peptides/administration & dosage , Peritoneal Neoplasms/drug therapy , Animals , Bacteriophages/genetics , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Disease Models, Animal , Drug Delivery Systems , Humans , Hyaluronan Receptors/genetics , Mice , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasm Metastasis , Peptides/genetics , Peritoneal Cavity/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology
3.
Cancer Sci ; 108(7): 1493-1503, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28444967

ABSTRACT

Tumor metastasis occurs naturally in pancreatic cancer, and the efficacy of chemotherapy is usually poor. Precision medicine, combining downregulation of target genes with chemotherapy drugs, is expected to improve therapeutic effects. Therefore, we developed a combined therapy of microRNA-21 antisense oligonucleotides (ASO-miR-21) and gemcitabine (Gem) using a targeted co-delivery nanoparticle (NP) carrier and investigated the synergistic inhibitory effects on pancreatic cancer cells metastasis and growth. Polyethylene glycol-polyethylenimine-magnetic iron oxide NPs were used to co-deliver ASO-miR-21 and Gem. An anti-CD44v6 single-chain variable fragment (scFvCD44v6 ) was used to coat the particles to obtain active and targeted delivery. Our results showed that the downregulation of the oncogenic miR-21 by ASO resulted in upregulation of the tumor-suppressor genes PDCD4 and PTEN and the suppression of epithelial-mesenchymal transition, which inhibited the proliferation and induced the clonal formation, migration, and invasion of pancreatic cancer cells in vitro. The co-delivery of ASO-miR-21 and Gem induced more cell apoptosis and inhibited the growth of pancreatic cancer cells to a greater extent than single ASO-miR-21 or Gem treatment in vitro. In animal tests, more scFvCD44v6 -PEG-polyethylenimine/ASO-magnetic iron oxide NP/Gem accumulated at the tumor site than non-targeted NPs and induced a potent inhibition of tumor proliferation and metastasis. Magnetic resonance imaging was used to observed tumor homing of NPs. These results imply that the combination of miR-21 gene silencing and Gem therapy using an scFv-functionalized NP carrier exerted synergistic antitumor effects on pancreatic cancer cells, which is a promising strategy for pancreatic cancer therapy.


Subject(s)
Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , MicroRNAs/antagonists & inhibitors , Molecular Targeted Therapy/methods , Oligonucleotides, Antisense/administration & dosage , Pancreatic Neoplasms/pathology , Animals , Deoxycytidine/administration & dosage , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Female , Ferric Compounds , Humans , Hyaluronan Receptors/administration & dosage , Metal Nanoparticles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Nanomedicine/methods , Polyethylene Glycols , Polyethyleneimine , Precision Medicine/methods , Single-Chain Antibodies/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine
4.
Lima; IETSI; 19 mayo 2016.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-910113

ABSTRACT

INTRODUCCIÓN: El presente informe está referido al Requerimiento de la Red Asistencial Arequipa de Especificaciones Técnicas de Implante de Hilano GF 20 y desbloqueo de Código SAP. ANTECEDENTES: -Resolución de Presidencia Ejecutiva Nº 152-PE-ESSALUD-2015, de fecha 23 de febrero del 2015, el Reglamento de Organización y Funciones del Instituto de Evaluación de Tecnologías en Salud e Investigación (IETSI); -Con Carta Nº 1028-G-RAAR-ESSALUD-2016, de fecha 11 de Abril del 2016, el Gerente Red Asistencial Arequipa - EsSalud, solicita a IETSI (Instituto de Evaluación de Tecnologías en Salud e Investigación), Especificaciones Técnicas de Implante de Hilano y desbloqueo de Código SAP. METODOLOGIA: Fuente Primaria: Los usuarios (Jefes del Servicio de Ortopedia y traumatología), han remitido su requerimiento para solicitar las especificaciones técnicas del implante Hilano GF20, por medio de carta Nº 1028-G-RAAR-ESSALUD-2016, de fecha 11 de Abril del 2016, por cuanto indican: Que en viscosuplementación está recomendada en las guías de tratamiento de Osteoartritis de las principales instituciones internacionales; Que el viscosuplemento de mayor similitud al líquido sinovial normal; Ha demostrado ser eficaz en diversos grados de steoartritis; Ha demostrado disminuir el dolor y mejorar la función articular en pacientes con osteoartritis Grado II y III; Ha demostrado retrasar la necesidad de reemplazo total de rodilla en una mediana de 2,1 años; En tratamiento de osteoartritis significó un ahorro de costos por paciente de 33% en el primer año, 10% y 9% en el segundo y tercer año. Asimismo se menciona como sustento técnico del requerimiento las Guías de Instituciones internacionales: OARSI (Osteoarthritis Research Society International), Guías de consenso de expertos basados en la evidencia del año 2007,y guías mexicanas de tratamiento de osteoartritis Rev. 2013, en las cuales se detalla que el uso de Hilano se encuentra en controversia. Búsqueda de Información: Se realizó una revisión sistemática de la evidencia científica sobre la eficacia, seguridad, efectividad relacionado al implante de Hilano. Se consultaron las siguientes bases de datos electrónicas: MEDLINE y PreMedline (1950-abril 2013), EMBASE (1980-abril 2013), PsycInfo (1887- abril 2013), CINAHL (1982- abril 2013), Cochrane Library (1982- abril 2013) y Centre for Reviews & Dissemination (1973- abril 2013). El análisis bibliográfico se complementó con consultas manuales, web de organizaciones y agencias de evaluaciones nacionales e internacionales. CONCLUSIONES: Una intervención debe primero mostrar tener un beneficio clínico, y el mejor comparador para mostrar esto sería el placebo, con el fin de identificar la magnitud del efecto sobre la respuesta al placebo. Solo si se prueba un efecto sobre placebo, se deben considerar la evidencia de sus comparaciones con otros tratamientos activos Los estudios que han evaluado la eficacia de Hilano GF20 relativo a placebo o suero salino, sugieren que Hilano GF20 reduce el dolor medido con las escalas de WOMAC hasta los tres meses, pero es semejante al placebo cuando el dolor es medido con la escala VAS a los tres y 6 meses. Además, después de tres meses, las reducciones importantes en el dolor se mostraron cuando se utilizaron múltiples inyecciones, pero este efecto no fue demostrado para inyecciones únicas y siempre que el dolor fuera medido con las escalas de WOMAC. Nuevamente, tras un periodo de tres meses, Hilano GF 20 y placebo fueron similares en su efecto en el dolor medido con la escala VAS. El probable efecto de Hilano GF 20 sobre el dolor no es consistente en función a la escala utilizada para medir el dolor de la articulación. La calidad de la evidencia que respalda estos probables efectos de beneficio de Hilano GF20 es de calidad baja o muy baja, lo cual implica que la confianza en los resultados es baja. El verdadero valor del efecto puede ser sustancialmente diferente al presentado, de realizarse estudios de adecuado diseño y conducción. El bloqueo de este Dispositivo se realiza hasta que se demuestre el costo-beneficio y beneficio clínico que representa a la Institución.


Subject(s)
Humans , Hyaluronan Receptors/administration & dosage , Prostheses and Implants , Viscoelastic Substances/administration & dosage , Cost-Benefit Analysis , Technology Assessment, Biomedical
5.
Vopr Onkol ; 62(3): 514-8, 2016.
Article in Russian | MEDLINE | ID: mdl-30463110

ABSTRACT

In this work the effect of RHAMM (receptor hyaluronan-mediated motility)-target peptides was investigated on the viability, apoptosis and necrosis of prostate cancer cells (PC3m-LN4). It has been established that RHAMM-target peptides inhibited on 90 % cell viability of PC3m-LN4 cells at a concentration of 10 ug / ml (2х10-7 M) for 48 h. It has shown that RHAMM-target peptides induced apoptosis and inhibited necrosis of tumor cells. RHAMM-target peptide had no effect on fibroblasts (non-tumor cells) and fibroblasts (RHAMM-/-). The studies also revealed that RHAMM-target peptides enhanced activity of caspase-3/7 in cancer cells.


Subject(s)
Cell Proliferation/drug effects , Extracellular Matrix Proteins/genetics , Hyaluronan Receptors/genetics , Peptides/administration & dosage , Prostatic Neoplasms/genetics , Apoptosis/drug effects , Caspase 3/genetics , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Matrix Proteins/administration & dosage , Fibroblasts/drug effects , Humans , Hyaluronan Receptors/administration & dosage , Male , Peptides/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Transfection
6.
BioDrugs ; 28(4): 393-402, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24567264

ABSTRACT

BACKGROUND AND OBJECTIVES: The aim of this study was to increase the serum half-life of recombinant CD44 hyaluronan (HA) binding domain by PEGylation. We have previously found that recombinant soluble CD44 HA binding domain (CD44HABD) and its non-HA-binding triple mutant CD44HABD(R41AY78SY79S) (CD44-3MUT) inhibits angiogenesis and subcutaneous tumor growth. However, this ~12 kDa recombinant protein displays a high serum clearance rate. METHODS: Here, we report the purification of monomeric CD44-3MUT from urea solubilized inclusion bodies using weak anion exchange chromatography and gel filtration. To increase the serum residence time of CD44-3MUT we PEGylated the resulting protein using 20 kDa methoxy-PEG-propionaldehyde. RESULTS: PEGylation of CD44-3MUT prolonged its in vivo serum half-life about 70-fold from 0.03 to 1.8 hours. Along with extended plasma residence time, PEGylation also increased the systemic exposure. By cell impedance assay we confirmed that PEGylated CD44-3MUT maintained its in vitro function. The results from the impedance assay additionally demonstrate that the CD44-3MUT effect on endothelial cells is mediated by vimentin. CONCLUSIONS: In summary, we have developed a purification protocol for large-scale production of CD44-3MUT and generated a PEGylated form of CD44-3MUT. HA binding domain of CD44(CD44HABD) and its modified non-HA binding form (CD44-3MUT) inhibit angiogenesis and tumor growth in vivo without disturbing HA-binding functions. CD44-3MUT has been PEGylated for use as a new type of anti-angiogenic human drug. PEGylation of CD44-3MUT improved pharmacokinetic properties but retains its functional activity.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Polyethylene Glycols/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Female , Half-Life , Hyaluronan Receptors/administration & dosage , Mice , Protein Binding , Rats , Rats, Inbred F344 , Recombinant Proteins/metabolism , Vimentin/metabolism
7.
J Drug Target ; 20(7): 561-73, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22758394

ABSTRACT

CD44 or hyaluronan receptor is a transmembrane receptor associated with aggressive tumour growth, proliferation, and metastasis. In normal physiology, this receptor has a crucial role in cell adhesion, inflammation, and repair processes. However, many tumour cells over-express this receptor and abuse it to become progressive and perpetual units. The article comments from common functioning of the CD44 receptor, to its diabolic multi-dimensional effects in promotion of malignant cells. It also illuminates the relations of CD44 endorsed processes with other biomolecular events in cancer progression. In an end, the review focuses comprehensively at ongoing researches to exploit the CD44 over-expression as a probable target in treatment, management, and diagnosis of malignancy.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Hyaluronan Receptors/physiology , Neoplasms/physiopathology , Animals , Disease Progression , Drug Carriers/administration & dosage , Drug Resistance, Multiple/physiology , Drug Screening Assays, Antitumor/methods , Humans , Hyaluronan Receptors/administration & dosage , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/physiology , Hyaluronoglucosaminidase/therapeutic use , Models, Biological , Nanoconjugates/administration & dosage , Neoplasms/drug therapy , Protein Isoforms/administration & dosage , Protein Isoforms/physiology
8.
Haematologica ; 95(7): 1191-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20081055

ABSTRACT

BACKGROUND: Recently, we demonstrated immunological and clinical responses to a RHAMM-R3 peptide vaccine in patients with acute myeloid leukemia, myelodysplastic syndrome and multiple myeloma. To improve the outcome of the vaccine, a second cohort was vaccinated with a higher dose of 1,000 microg peptide. DESIGN AND METHODS: Nine patients received four vaccinations subcutaneously at a biweekly interval. Immunomonitoring of cytotoxic CD8(+) as well as regulatory CD4(+) T cells was performed by flow cytometry as well as by enzyme-linked immunospot (ELISpot) assays. Parameters of clinical response were assessed. RESULTS: In 4 of 9 patients (44%) we detected positive immunological responses. These patients showed an increase of CD8(+)RHAMM-R3_tetramer(+)/CD45RA(+)/CCR7(-)/CD27(-)/CD28(-) effector T cells and an increase of R3-specific CD8+ T cells. Two of these patients showed a significant decrease of regulatory T cells (Tregs). In one patient without response Tregs frequency increased from 5 to 16%. Three patients showed clinical effects: one patient with myelodysplastic syndrome RAEB-1 showed a reduction of leukemic blasts in the bone marrow, another myelodysplastic syndrome patient an improvement of peripheral blood counts and one patient with multiple myeloma a reduction of free light chains. Clinical and immunological reactions were lower in this cohort than in the 300 microg cohort. CONCLUSIONS: High-dose RHAMM-R3 peptide vaccination induced immunological responses and positive clinical effects. Therefore, RHAMM constitutes a promising structure for further targeted immunotherapies in patients with different hematologic malignancies. However, higher doses of peptide did not improve the frequency and intensity of immune responses in this trial.


Subject(s)
Cancer Vaccines/administration & dosage , Extracellular Matrix Proteins/administration & dosage , Hyaluronan Receptors/administration & dosage , Immunity/drug effects , Peptide Fragments/administration & dosage , Aged , Cancer Vaccines/chemistry , Dose-Response Relationship, Drug , Extracellular Matrix Proteins/immunology , Female , Humans , Hyaluronan Receptors/immunology , Male , Middle Aged , Peptide Fragments/immunology , T-Lymphocytes/immunology , Vaccination
9.
J Immunol ; 181(7): 4676-84, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802070

ABSTRACT

Cachexia is the dramatic weight loss and muscle atrophy seen in chronic disease states, including autoimmunity, cancer, and infection, and is often associated with lymphopenia. We have previously shown that CD4(+) T cells that express the lowest density of CD44 (CD4(+)CD44(v.low)) are significantly reduced in diabetic NOD mice that are cachexic compared with diabetic mice that are not cachexic. Using this model, and a model of cancer cachexia, we test the hypothesis that CD4(+)CD44(v.low) cells play an active role in protecting the host from cachexia. CD4(+)CD44(v.low) cells, but not CD4(+) cells depleted of CD44(v.low) cells, delay the onset of wasting when infused into either diabetic or prediabetic NOD recipients. However, no significant effect on the severity of diabetes was detected. In a model of cancer cachexia, they significantly reduce muscle atrophy, and inhibit muscle protein loss and DNA loss, even when given after the onset of cachexia. Protection from wasting and muscle atrophy by CD4(+)CD44(v.low) cells is associated with protection from lymphopenia. These data suggest, for the first time, a role for an immune cell subset in protection from cachexia, and further suggest that the mechanism of protection is independent of protection from autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cachexia/immunology , Cachexia/therapy , Animals , CD4-Positive T-Lymphocytes/metabolism , Cachexia/prevention & control , Carcinoma, Lewis Lung , Cell Line, Tumor , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/immunology , Female , Hyaluronan Receptors/administration & dosage , Hyaluronan Receptors/biosynthesis , Incidence , Lymphopenia/immunology , Lymphopenia/therapy , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Muscular Atrophy/immunology , Muscular Atrophy/prevention & control , Neoplasm Transplantation , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Wasting Syndrome/immunology , Wasting Syndrome/therapy
10.
Biochem Biophys Res Commun ; 343(2): 609-16, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16554035

ABSTRACT

Bone marrow stromal cells produce large amounts of extracellular matrix and cytokines. Amongst them, hyaluronan, a glycosaminoglycan and ligand for the cell surface molecule CD44, and TGFbeta1, a cytokine particularly important in monocyte differentiation. We have studied in vitro the role of hyaluronan and TGFbeta1 in the differentiation process of U937 monocytic progenitor cells. We provide evidence that, in the presence of whole blood-derived serum, the addition of hyaluronan is sufficient to induce the expression of NADPH-oxidase components but not of other monocytic markers (CD14, CD11b, and VLA-4). In the presence of plasma-derived serum, besides hyaluronan, the additional presence of TGFbeta1 was required for the expression of all of the components of the NADPH oxidase. We further show that hyaluronan mediates its effect through CD44. We conclude that cell matrix factors act cooperatively with cytokines to induce the expression of the components of the NADPH-oxidase in monocytic progenitor cells.


Subject(s)
Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/enzymology , Hyaluronan Receptors/administration & dosage , Monocytes/cytology , Monocytes/enzymology , NADPH Oxidases/metabolism , Transforming Growth Factor beta/administration & dosage , Cell Differentiation/drug effects , Cell Line , Dose-Response Relationship, Drug , Drug Combinations , Drug Synergism , Gene Expression Regulation, Enzymologic/drug effects , Granulocyte Precursor Cells/drug effects , Humans , Monocytes/drug effects , Transforming Growth Factor beta1
11.
J Immunol ; 163(3): 1619-27, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10415067

ABSTRACT

At sites of chronic inflammation seen during infections, autoimmunity, graft-vs-host response, and cytokine therapy, endothelial cell injury is known to occur, the exact mechanism of which is unknown. In the current study we used IL-2-induced vascular leak syndrome (VLS) as a model to investigate whether cytotoxic lymphocytes use CD44 in mediating endothelial cell injury. Administration of IL-2 to wild-type mice triggered significant VLS in the lungs and liver. In contrast, in CD44 knockout (KO) mice, IL-2-induced VLS was markedly reduced in the lungs and liver. IL-2-treated wild-type and CD44 KO mice had similar levels of perivascular infiltration with lymphocytes in the lungs and liver. This suggested that the decrease in VLS seen in CD44 KO mice was not due to the inability of lymphocytes to migrate to these organs. Ultrastructural studies demonstrated extensive endothelial cell damage in the lungs and liver of IL-2-treated wild-type, but not CD44 KO, mice. Moreover, CD44-KO mice exhibited a marked decrease in IL-2-induced lymphokine-activated killer cell activity. The induction of VLS was dependent on the expression of CD44 on immune cells rather than endothelial cells because adoptive transfer of CD44+, but not CD44- spleen cells along with IL-2 into CD44 KO mice triggered VLS. The IL-2-induced VLS was blocked by administration of F(ab')2 of Abs against CD44. The current study demonstrates that CD44 plays a key role in endothelial cell injury. Blocking CD44 in vivo may offer a novel therapeutic approach to prevent endothelial cell injury by cytotoxic lymphocytes in a variety of clinical disease models.


Subject(s)
Capillary Leak Syndrome/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Hyaluronan Receptors/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Capillary Leak Syndrome/etiology , Capillary Leak Syndrome/genetics , Capillary Leak Syndrome/pathology , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Endothelium, Vascular/chemistry , Female , Hyaluronan Receptors/administration & dosage , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/genetics , Hyaluronic Acid/pharmacology , Immunoglobulin Fab Fragments/administration & dosage , Injections, Intraperitoneal , Interleukin-2/administration & dosage , Killer Cells, Lymphokine-Activated/immunology , Liver/pathology , Liver/ultrastructure , Lung/pathology , Lung/ultrastructure , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/cytology , Spleen/transplantation , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...