Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Control Release ; 328: 503-513, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32860926

ABSTRACT

Liposome-supported peritoneal dialysis (LSPD) with transmembrane pH-gradient liposomes was previously shown to enhance ammonia removal in cirrhotic rats and holds promise for the treatment of hyperammonemic crises-associated disorders. The main objective of this work was to conduct the preclinical evaluation of LSPD in terms of pharmacokinetics, ammonia uptake, and toxicology to seek regulatory approval for a first-in-human study. The formulation containing citric acid-loaded liposomes was administered intraperitoneally at two different doses once daily for ten days to healthy minipigs. It was also tested in a domestic pig model of hyperammonemia. The pharmacokinetics of citric acid and 1,2-dipalmitoyl-sn-glycero-3-phosphocholine was linear following intraperitoneal administration of medium and high dose. There was no systemic accumulation following daily doses over ten days. The systemic exposure to phospholipids remained low. Furthermore, the liposome-containing peritoneal fluid contained significantly higher ammonia levels than the liposome-free control, demonstrating efficient ammonia sequestration in the peritoneal space. This was indeed confirmed by the ability of LSPD to decrease plasmatic ammonia levels in artificially induced hyperammonemic pigs. LSPD was well tolerated, and no complement activation-related pseudoallergy reactions were observed. The safety profile, the linear pharmacokinetics of citric acid following repeated administrations of LSPD as well as the linear dose-dependent ammonia sequestration in the peritoneal space provide a strong basis for the clinical investigation of LSPD.


Subject(s)
Hyperammonemia , Peritoneal Dialysis , Ammonia , Animals , Ascitic Fluid , Hyperammonemia/drug therapy , Liposomes , Rats , Swine , Swine, Miniature
2.
Anal Bioanal Chem ; 410(21): 5245-5253, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29947896

ABSTRACT

Asymmetric flow field-flow fractionation (AF4) coupled with UV-Vis spectroscopy, multi-angle light scattering (MALS) and refractive index (RI) detection has been applied for the characterization of MIL-100(Fe) nanoMOFs (metal-organic frameworks) loaded with nucleoside reverse transcriptase inhibitor (NRTI) drugs for the first time. Empty nanoMOFs and nanoMOFs loaded with azidothymidine derivatives with three different degrees of phosphorylation were examined: azidothymidine (AZT, native drug), azidothymidine monophosphate (AZT-MP), and azidothymidine triphosphate (AZT-TP). The particle size distribution and the stability of the nanoparticles when interacting with drugs have been determined in a time frame of 24 h. Main achievements include detection of aggregate formation in an early stage and monitoring nanoMOF morphological changes as indicators of their interaction with guest molecules. AF4-MALS proved to be a useful methodology to analyze nanoparticles engineered for drug delivery applications and gave fundamental data on their size distribution and stability. Graphical abstract ᅟ.


Subject(s)
Anti-HIV Agents/administration & dosage , Coordination Complexes/chemistry , Drug Carriers/chemistry , Metal-Organic Frameworks/chemistry , Nanoparticles/chemistry , Zidovudine/administration & dosage , Anti-HIV Agents/chemistry , Antimetabolites/administration & dosage , Antimetabolites/chemistry , Dideoxynucleotides/administration & dosage , Dideoxynucleotides/chemistry , Dynamic Light Scattering , Fractionation, Field Flow , Models, Molecular , Particle Size , Refractometry , Spectrophotometry, Ultraviolet , Thymine Nucleotides/administration & dosage , Thymine Nucleotides/chemistry , Zidovudine/analogs & derivatives , Zidovudine/chemistry
3.
J Control Release ; 278: 57-65, 2018 05 28.
Article in English | MEDLINE | ID: mdl-29601930

ABSTRACT

Peritoneal dialysis (PD) performed with transmembrane pH-gradient liposomes was reported to efficiently remove ammonia from the body, representing a promising alternative to current standard-of-care for patients with severe hepatic encephalopathy. In this study, we further characterized the properties of liposome-supported peritoneal dialysis (LSPD) by 1) assessing its in-use stability in the presence of ascitic fluids from liver-disease patients; 2) investigating its interactions with drugs that are commonly administered to acute-on-chronic liver failure patients; and 3) analyzing the in vivo extraction profile of LSPD. We found that LSPD fluid maintained its in vitro ammonia uptake capability when combined with ascitic fluids. The co-incubation of selected drugs (e.g., beta-blockers, antibiotics, diuretics) with LSPD fluids and ammonia resulted in limited interaction effects for most compounds except for two fluoroquinolones and propranolol. However, considering the experimental set-up, these results should be interpreted with caution and confirmatory drug-drug interaction studies in a clinical setting will be required. Finally, metabolite-mapping analysis on dialysates of LSPD-treated rats revealed that the liposomes did not remove important metabolites more than a conventional PD fluid. Overall, these findings confirm that LSPD is a potentially safe and effective approach for treating hyperammonemic crises in the context of acute-on-chronic liver failure.


Subject(s)
Ammonia/metabolism , Ascitic Fluid/metabolism , Hyperammonemia/therapy , Peritoneal Dialysis/methods , Acute-On-Chronic Liver Failure/drug therapy , Acute-On-Chronic Liver Failure/metabolism , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/metabolism , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/metabolism , Diuretics/administration & dosage , Diuretics/metabolism , Humans , Hydrogen-Ion Concentration , Hyperammonemia/physiopathology , Liposomes , Male , Rats , Rats, Sprague-Dawley , Severity of Illness Index
4.
Sci Rep ; 7(1): 8353, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827534

ABSTRACT

Positively charged cyclodextrins (PCCDs) are molecular carriers of particular interest for their ability to readily enter into cancer cells. Of main interest, guanidino- and aminoalkyl- PCCDs can be conveniently synthesized and form stable and strong inclusion complexes with various active molecules bearing phosphate groups. We have addressed here the challenge to deliver into cancer cells phosphorylated gemcitabine drugs well known for their instability and inability to permeate cell membranes. NMR data corroborated by semiempirical theoretical calculations have shown that aminoalkyl-CDs form sufficiently stable complexes with both mono- and tri-phosphate forms of gemcitabine by simple mixing of the compounds in aqueous solution at physiological pH. Confocal microscopy and radioactivity counting experiments revealed that the developed systems enabled phosphorylated gemcitabine to penetrate efficiently into aggressive human breast cancer cells (MCF7), eventually leading to a substantial reduction of IC50 values. Moreover, compared to free drugs, phosphorylated metabolites of gemcitabine encapsulated in PCCDs displayed improved in vitro activities also on the aggressive human cancer cells CCRF-CEM Ara-C/8 C, a nucleoside transport-deficient T leukemia cell line. The current study offers the proof-of-principle that phosphorylated nucleoside drugs could be efficiently transported by PCCDs into cancer cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Breast Neoplasms/metabolism , Cyclodextrins/metabolism , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Lymphoma, T-Cell/metabolism , Biological Transport , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cyclodextrins/chemistry , Cytarabine/pharmacology , Deoxycytidine/pharmacology , Female , Humans , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/pathology , Models, Theoretical , Phosphorylation , Tumor Cells, Cultured , Gemcitabine
5.
Biochim Biophys Acta Gen Subj ; 1861(6): 1606-1616, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28137620

ABSTRACT

BACKGROUND: Metal-organic framework nanoparticles (nanoMOFs) are biodegradable highly porous materials with a remarkable ability to load therapeutic agents with a wide range of physico-chemical properties. Engineering the nanoMOFs surface may provide nanoparticles with higher stability, controlled release, and targeting abilities. Designing postsynthetic, non-covalent self-assembling shells for nanoMOFs is especially appealing due to their simplicity, versatility, absence of toxic byproducts and minimum impact on the original host-guest ability. METHODS: In this study, several ß-cyclodextrin-based monomers and polymers appended with mannose or rhodamine were randomly phosphorylated, and tested as self-assembling coating building blocks for iron trimesate MIL-100(Fe) nanoMOFs. The shell formation and stability were studied by isothermal titration calorimetry (ITC), spectrofluorometry and confocal imaging. The effect of the coating on tritium-labeled AZT-PT drug release was estimated by scintillation counting. RESULTS: Shell formation was conveniently achieved by soaking the nanoparticles in self-assembling agent aqueous solutions. The grafted phosphate moieties enabled a firm anchorage of the coating to the nanoMOFs. Coating stability was directly related to the density of grafted phosphate groups, and did not alter nanoMOFs morphology or drug release kinetics. CONCLUSION: An easy, fast and reproducible non-covalent functionalization of MIL-100(Fe) nanoMOFs surface based on the interaction between phosphate groups appended to ß-cyclodextrin derivatives and iron(III) atoms is presented. GENERAL SIGNIFICANCE: This study proved that discrete and polymeric phosphate ß-cyclodextrin derivatives can conform non-covalent shells on iron(III)-based nanoMOFs. The flexibility of the ß-cyclodextrin to be decorated with different motifs open the way towards nanoMOFs modifications for drug delivery, catalysis, separation, imaging and sensing. This article is part of a Special Issue entitled "Recent Advances in Bionanomaterials" Guest Editors: Dr. Marie-Louise Saboungi and Dr. Samuel D. Bader.


Subject(s)
Click Chemistry/methods , Iron Compounds/chemistry , Magnetics/methods , Mannose/chemistry , Metal Nanoparticles/chemistry , Nanotechnology/methods , Rhodamines/chemistry , Zidovudine/chemistry , beta-Cyclodextrins/chemistry , Calorimetry/methods , Drug Liberation , Kinetics , Mannose/analogs & derivatives , Microscopy, Confocal , Phosphorylation , Porosity , Solubility , Spectrometry, Fluorescence , Surface Properties , Thermodynamics
6.
J Drug Target ; 23(7-8): 759-67, 2015.
Article in English | MEDLINE | ID: mdl-26453171

ABSTRACT

Metal-organic frameworks (MOFs) are coordination polymers of interest for biomedical applications. Of particular importance, nanoparticles made of iron(III) trimesate (MIL-100, MIL standing for Material Institut Lavoisier) (nanoMOFs) can be conveniently synthesised under mild and green conditions. They were shown to be biodegradable, biocompatible and efficient to encapsulate a variety of active molecules. We have addressed here the challenges to encapsulate a highly hydrophilic anticancer prodrug, phosphated gemcitabin (Gem-MP) known for its instability and inability to bypass cell membranes. MIL-100 nanoMOFs acted as efficient "nanosponges", soaking Gem-MP from its aqueous solution with almost perfect efficiency (>98%). Maximal loadings reached ∼30 wt% reflecting the strong interaction between the drug and the iron trimesate matrices. Neither degradation nor loss of crystalline structure was observed after the loading process. Storage of the loaded nanoMOFs in water did not result in drug release over three days. However, Gem-MP was released in media containing phosphates, as a consequence to particle degradation. Drug-loaded nanoMOFs were effective against pancreatic PANC-1 cells, in contrast to free drug and empty nanoMOFs. However, an efflux phenomenon could contribute to reduce the efficacy of the nanocarriers. Size optimization and surface modification of the nanoMOFs are expected to further improve these findings.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Metal Nanoparticles , Pancreatic Neoplasms/drug therapy , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Crystallization , Deoxycytidine/administration & dosage , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Liberation , Drug Stability , Drug Storage , Ferric Compounds/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Organometallic Compounds/chemistry , Pancreatic Neoplasms/pathology , Phosphates/chemistry , Polymers/chemistry , Prodrugs/administration & dosage , Prodrugs/chemistry , Gemcitabine
7.
J Phys Chem B ; 118(29): 8532-9, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24960194

ABSTRACT

Doxorubicin (DOX) entrapment in porous Fe(III)-trimesate metal organic frameworks (MIL-100(Fe)) nanoparticles was investigated in neutral Tris buffer via UV-vis absorption, circular dichroism (CD), and fluorescence. The binding constants and the absolute spectra of the DOX-MIL-100(Fe) complexes were determined via absorption and fluorescence titrations. A binding model where DOX associates as monomer to the dehydrated Fe3O (OH)(H2O)2 [(C6H3)(CO2)3]2 structural unit in 1:1 stoichiometry, with apparent association constant of (1.1 to 1.8) × 10(4) M(-1), was found to reasonably fit the experimental data. Spectroscopic data indicate that DOX binding occurs via the formation of highly stable coordination bonds between one or both deprotonated hydroxyl groups of the aglycone moiety and coordinatively unsaturated Fe(III) centers. Complete quenching of the DOX fluorescence and remarkable thermal and photochemical stability were observed for DOX incorporated in the MIL-100(Fe) framework.


Subject(s)
Doxorubicin/chemistry , Drug Carriers/chemistry , Iron/chemistry , Nanoparticles/chemistry , Organometallic Compounds/chemistry , Drug Stability , Free Radicals/chemistry , Models, Molecular , Molecular Conformation , Porosity
8.
J Med Chem ; 57(2): 411-20, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24345217

ABSTRACT

An essential challenge in the development of nanosized metal organic framework (nanoMOF) materials in biomedicine is to develop a strategy to stabilize their supramolecular structure in biological media while being able to control drug encapsulation and release. We have developed a method to efficiently encapsulate topotecan (TPT, 1), an important cytotoxic drug, in biodegradable nanoMOFs. Once inside the pores, 1 monomers aggregate in a "ship in a bottle" fashion, thus filling practically all of the nanoMOFs' available free volume and stabilizing their crystalline supramolecular structures. Highly efficient results have been found with the human pancreatic cell line PANC1, in contrast with free 1. We also demonstrate that one- and two-photon light irradiation emerges as a highly promising strategy to promote stimuli-dependent 1 release from the nanoMOFs, hence opening new standpoints for further developments in triggered drug delivery.


Subject(s)
Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Light , Nanoparticles/chemistry , Topotecan/chemistry , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Compounding , Ferric Compounds/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Photons , Porosity , Topotecan/administration & dosage , Tricarboxylic Acids/chemistry , Tumor Protein, Translationally-Controlled 1
9.
Adv Healthc Mater ; 2(12): 1630-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23776182

ABSTRACT

Nanoscale mesoporous iron carboxylates metal-organic frameworks (nanoMOFs) have recently emerged as promising platforms for drug delivery, showing biodegradability, biocompatibility and important loading capability of challenging highly water-soluble drugs such as azidothymidine tryphosphate (AZT-TP). In this study, nanoMOFs made of iron trimesate (MIL-100) were able to act as efficient molecular sponges, quickly adsorbing up to 24 wt% AZT-TP with entrapment efficiencies close to 100%, without perturbation of the supramolecular crystalline organization. These data are in agreement with molecular modelling predictions, indicating maximal loadings of 33 wt% and preferential location of the drug in the large cages. Spectrophotometry, isothermal titration calorimetry, and solid state NMR investigations enable to gain insight on the mechanism of interaction of AZT and AZT-TP with the nanoMOFs, pointing out the crucial role of phosphates strongly coordinating with the unsaturated iron(III) sites. Finally, contrarily to the free AZT-TP, the loaded nanoparticles efficiently penetrate and release their cargo of active triphosphorylated AZT inside major HIV target cells, efficiently protecting against HIV infection.


Subject(s)
Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/chemistry , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Nanocomposites/chemistry , Anti-Retroviral Agents/pharmacokinetics , Cells, Cultured , Dideoxynucleotides/administration & dosage , Dideoxynucleotides/chemistry , Dideoxynucleotides/pharmacokinetics , Ferric Compounds/pharmacokinetics , HIV-1/drug effects , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Nanocomposites/administration & dosage , Thymine Nucleotides/administration & dosage , Thymine Nucleotides/chemistry , Thymine Nucleotides/pharmacokinetics , Zidovudine/administration & dosage , Zidovudine/analogs & derivatives , Zidovudine/chemistry , Zidovudine/pharmacokinetics
10.
J Immunother ; 36(3): 181-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23502765

ABSTRACT

T lymphocytes engineered to express chimeric antigen receptors (CARs) interact directly with cell surface molecules, bypassing MHC antigen presentation dependence. We generated human anti-CD19ζ CAR cytotoxic T lymphocytes and cytokine-induced killer cells and studied their sensitivity to the expression of adhesion molecules for the killing of primary B-lineage acute lymphoblastic leukemia (B-ALL) targets. Despite a very low basal expression of surface adhesion molecules, B-ALL blasts were lysed by the anti-CD19ζ-CAR transduced effectors as expected. We next investigated the regulatory role of adhesion molecules during CAR-mediated cytolysis. The blocking of these accessory molecules strongly limited the chimeric effector's cytotoxicity. Thereafter, B-ALL cells surface adhesion molecule level expression was induced by IFN-γ or by the combined use of CD40L and IL-4 and the cells were submitted to anti-CD19ζ-CAR transduced effectors lysis. Upregulation of adhesion molecules expression by blasts potentiated their killing. The improved cytotoxicity observed was dependent on target surface expression of adhesion molecules, particularly CD54. Taken together, these results indicate that adhesion molecules, and principally CD54, are involved in the efficiency of recognition by effector chimeric ζ. These observations have potential implications for the design of immunotherapy treatment approaches for hematological malignancies and tumors based on the adoption of CAR effector cells.


Subject(s)
Antigens, CD19/immunology , Intercellular Adhesion Molecule-1/metabolism , Leukemia, B-Cell/immunology , Receptors, Antigen, B-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Burkitt Lymphoma/immunology , CD40 Antigens/metabolism , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Humans , Intercellular Adhesion Molecule-1/genetics , Interferon-gamma/immunology , Interleukin-4/metabolism , Jurkat Cells , Leukemia, B-Cell/metabolism , Leukocytes, Mononuclear/immunology , RNA Interference , RNA, Small Interfering , Receptors, Antigen, B-Cell/biosynthesis , Up-Regulation
11.
J Mater Chem B ; 1(34): 4231-4242, 2013 Sep 14.
Article in English | MEDLINE | ID: mdl-32261018

ABSTRACT

Encapsulation of azidothymidine (AZT) or its phosphorylated derivatives (AZT-MP and AZT-TP) has been performed using nanoparticles of the porous crystalline iron(iii) trimesate metal-organic framework MIL-100(Fe). The number of phosphate groups per nucleoside analogue has a high impact on the drug loading capacity, and their interaction with the Lewis acid sites from the nanoMOFs is also discussed through a combination of techniques such as UV-vis absorption, circular dichroism, isothermal titration calorimetry, HPLC and molecular simulations. Finally, the effect of the differences in terms of host-guest interactions is discussed through the release in physiological buffers of AZT, AZT-MP and AZT-TP. New perspectives for the nanoencapsulation of monophosphorylated nucleoside analogues for effective anti-cancer and anti-viral therapies are then discussed.

12.
Photochem Photobiol Sci ; 11(8): 1285-92, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22576059

ABSTRACT

The association of doxorubicin (DOX) and artemisinin (ART) to a ß-CyD-epichlorohydrin crosslinked polymer (pß-CyD), organized in nanoparticles of ca. 15 nm size, was investigated in neutral aqueous medium by circular dichroism (CD), UV-vis absorption and fluorescence. The stability constants and the absolute CD spectra of the drug complexes were determined by global analysis of multiwavelength data from spectroscopic titrations. The polymer pß-CyD proved able to disrupt the DOX dimer when the latter is the predominant form of DOX in solution. The spectroscopic and photophysical properties of the complexes evidenced an alcohol-like environment for ART and an improved inherent emission ability for DOX in the nanoparticle frame.


Subject(s)
Artemisinins/chemistry , Doxorubicin/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Spectrum Analysis , beta-Cyclodextrins/chemistry , Epichlorohydrin/chemistry , Models, Molecular , Molecular Conformation , Particle Size
13.
J Immunother ; 34(6): 469-79, 2011.
Article in English | MEDLINE | ID: mdl-21654519

ABSTRACT

The identification of the optimal T-cell effector subtype is a crucial issue for adoptive cell therapy with chimeric receptor-modified T cells. The ideal T cell population must be able to home toward tumor site, exert prolonged antitumoral activity, and display minimal toxicity against normal tissues. Therefore, we characterized the in vitro antitumoral properties of three effector T-cell populations: Epstein-Barr virus-specific cytotoxic T lymphocytes (EBV-CTLs), cytokine-induced killer (CIK) cells, and γ9δ2 T (GDT) cells, after transduction with a chimeric receptor specific for the CD33 antigen, broadly expressed on acute myeloid leukemia cells. EBV-CTLs, CIK, and GDT cells were generated and transduced with high efficiency with a retroviral vector coding for an anti-CD33-ζ chimeric receptor without alterations of their native phenotype. Anti-CD33-ζ chimeric receptor-redirected T cells displayed analogous in vitro chemotactic activity toward CXCL12. In addition, anti-CD33-ζ chimeric receptor-expressing EBV-CTLs, CIK, and GDT cells showed potent and similar cytotoxicity against several CD33⁺ leukemic targets both in short-term 4-hours-5¹chromium-release assays (mean killing vs primary leukemic cells at effector:target ratio of 5:1; 50%, 61%, and 50% for EBV-CTLs, CIK, and GDT cells, respectively) and in long-term assays, where they were cocultured with leukemic cells for 6 days on stromal mesenchymal cells (mean survival of primary leukemic cells at effector:target ratio of 1:100; 18%, 16%, and 29% for EBV-CTLs, CIK, and GDT cells, respectively). Moreover, all effector cells acquired consistent capability to proliferate in vitro after contact with CD33⁺ cells and to release high and comparable levels of immunostimulatory cytokines, while secreting similar low amount of immunoregulatory cytokines as the unmanipulated counterpart. Our results indicate that expression of an anti-CD33-ζ chimeric receptor potently and similarly increase the antileukemic functions of different effector T-cell subtypes, underlying the impossibility to identify a more potent T-cell population through in vitro analysis, and consistently with recent observations that have emerged from clinical trials with chimeric receptor-modified T cells, suggesting the need to perform such type of studies in the human setting.


Subject(s)
Cell- and Tissue-Based Therapy , Chimerism , Leukemia/immunology , Leukemia/therapy , Receptors, Antigen, T-Cell , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , CD3 Complex/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Line, Tumor , Chemotaxis, Leukocyte/immunology , Child , Genetic Vectors/genetics , HL-60 Cells , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/immunology , Humans , K562 Cells , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic
14.
Blood ; 117(18): 4736-45, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21406718

ABSTRACT

Chronic lymphocytic leukemia (CLL) is characterized by an accumulation of mature CD19(+)CD5(+)CD20(dim) B lymphocytes that typically express the B-cell activation marker CD23. In the present study, we cloned and expressed in T lymphocytes a novel chimeric antigen receptor (CAR) targeting the CD23 antigen (CD23.CAR). CD23.CAR(+) T cells showed specific cytotoxic activity against CD23(+) tumor cell lines (average lysis 42%) and primary CD23(+) CLL cells (average lysis 58%). This effect was obtained without significant toxicity against normal B lymphocytes, in contrast to CARs targeting CD19 or CD20 antigens, which are also expressed physiologically by normal B lymphocytes. Moreover, CLL-derived CD23.CAR(+) T cells released inflammatory cytokines (1445-fold more TNF-ß, 20-fold more TNF-α, and 4-fold more IFN-γ). IL-2 was also produced (average release 2681 pg/mL) and sustained the antigen-dependent proliferation of CD23.CAR(+) T cells. Redirected T cells were also effective in vivo in a CLL Rag2(-/-)γ(c)(-/-) xenograft mouse model. Compared with mice treated with control T cells, the infusion of CD23.CAR(+) T cells resulted in a significant delay in the growth of the MEC-1 CLL cell line. These data suggest that CD23.CAR(+) T cells represent a selective immunotherapy for the elimination of CD23(+) leukemic cells in patients with CLL.


Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Receptors, IgE/antagonists & inhibitors , Animals , B-Lymphocytes/immunology , Cell Line, Tumor , Coculture Techniques , Cytokines/biosynthesis , Cytotoxicity, Immunologic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression , Humans , Interleukin-2/biosynthesis , Lymphocyte Activation , Mice , Mice, Knockout , Receptors, IgE/genetics , Receptors, IgE/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
15.
Haematologica ; 95(12): 2144-52, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20713459

ABSTRACT

BACKGROUND: Cytokine-induced killer cells are ex vivo-expanded cells with potent antitumor activity. The infusion of cytokine-induced killer cells in patients with acute myeloid leukemia relapsing after allogeneic hematopoietic stem cell transplant is well tolerated, but limited clinical responses have been observed. To improve their effector functions against acute myeloid leukemia, we genetically modified cytokine-induced killer cells with chimeric receptors specific for the CD33 myeloid antigen. DESIGN AND METHODS: SFG-retroviral vectors coding for anti-CD33-ζ and anti-CD33-CD28-OX40-ζ chimeric receptors were used to transduce cytokine-induced killer cells. Transduced cells were characterized in vitro for their ability to lyse leukemic targets (4-hour (51)chromium-release and 6-day co-cultures assays on human stromal mesenchymal cells), to proliferate ((3)H-thymidine-incorporation assay) and to secrete cytokines (flow cytomix assay) after contact with acute myeloid leukemia cells. Their activity against normal CD34(+) hematopoietic progenitor cells was evaluated by analyzing the colony-forming unit capacity after co-incubation. RESULTS: Cytokine-induced killer cells were efficiently transduced with the anti-CD33 chimeric receptors, maintaining their native phenotype and functions and acquiring potent cytotoxicity (up to 80% lysis after 4-hour incubation) against different acute myeloid leukemia targets, as also confirmed in long-term killing experiments. Moreover, introduction of the anti-CD33 chimeric receptors was accompanied by prominent CD33-specific proliferative activity, with the release of high levels of immunostimulatory cytokines. The presence of CD28-OX40 in chimeric receptor endodomain was associated with a significant amelioration of the anti-leukemic activity of cytokine-induced killer cells. Importantly, even though the cytokine-induced killer cells transduced with anti-CD33 chimeric receptors showed toxicity against normal hematopoietic CD34(+) progenitor cells, residual clonogenic activity was preserved. CONCLUSIONS: Our results indicate that anti-CD33 chimeric receptors strongly enhance anti-leukemic cytokine-induced killer cell functions, suggesting that cytokine-induced killer cells transduced with these molecules might represent a promising optimized tool for acute myeloid leukemia immunotherapy.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cytokine-Induced Killer Cells/immunology , Leukemia, Myeloid/therapy , Acute Disease , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Child , Coculture Techniques , Cytokine-Induced Killer Cells/cytology , Cytokine-Induced Killer Cells/metabolism , Cytotoxicity, Immunologic/immunology , Flow Cytometry , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid/immunology , Leukemia, Myeloid/pathology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/immunology , Recombinant Fusion Proteins/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Time Factors
16.
J Biomed Biotechnol ; 2010: 234540, 2010.
Article in English | MEDLINE | ID: mdl-20617141

ABSTRACT

Adoptive transfer of specific cytotoxic T lymphocytes (CTL) and Cytokine Induced Killer Cells (CIK) following genetic engineering of T-cell receptor zeta hold promising perspective in immunotherapy. In the present work we focused on the mechanisms of anti-tumor action of effectors transduced with an anti-CD19 chimaeric receptor in the context of B-lineage acute lymphoblastic leukemia (B-ALL). Primary B-ALL blasts were efficiently killed by both z-CD19 CTL and z-CD19 CIK effectors. The use of death receptor mediated apoptosis of target cells was excluded since agonists molecules of Fas and TRAIL-receptors failed to induce cell death. Perforin/granzyme pathway was found to be the mechanism of chimaeric effectors mediated killing. Indeed, cytolytic effector molecules perforin as well as granzymes were highly expressed by CTL and CIK. CD19 specific stimulation of transduced effectors was associated with degranulation as attested by CD107 membrane expression and high IFN-gamma and TNF-alpha release. Moreover inhibitors of the perforin-based cytotoxic pathway, Ca(2+)-chelating agent EGTA and Concanamycin A, almost completely abrogated B-ALL blast killing. In conclusion we show that the cytolysis response of z-CD19 chimaeric effectors is predominantly mediated via perforin/granzyme pathway and is independent of death receptors signaling in primary B-ALL.


Subject(s)
Apoptosis , Cytokine-Induced Killer Cells/metabolism , Immunotherapy, Adoptive/methods , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes, Cytotoxic/metabolism , Antigens, CD19/metabolism , Cell Line , Cell Transformation, Neoplastic , Flow Cytometry , Granzymes/metabolism , Humans , Lysosomal Membrane Proteins/metabolism , Perforin/metabolism , Receptors, Death Domain/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...