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










Publication year range
1.
Cancer Cell ; 35(4): 649-663.e10, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30991025

ABSTRACT

The BCR-ABL1 fusion protein is the cause of chronic myeloid leukemia (CML) and of a significant fraction of adult-onset B cell acute lymphoblastic leukemia (B-ALL) cases. Using mouse models and patient-derived samples, we identified an essential role for γ-catenin in the initiation and maintenance of BCR-ABL1+ B-ALL but not CML. The selectivity was explained by a partial γ-catenin dependence of MYC expression together with the susceptibility of B-ALL, but not CML, to reduced MYC levels. MYC and γ-catenin enabled B-ALL maintenance by augmenting BIRC5 and enforced BIRC5 expression overcame γ-catenin loss. Since γ-catenin was dispensable for normal hematopoiesis, these lineage- and disease-specific features of canonical Wnt signaling identified a potential therapeutic target for the treatment of BCR-ABL1+ B-ALL.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Wnt Signaling Pathway , gamma Catenin/metabolism , Animals , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Survivin/genetics , Survivin/metabolism , beta Catenin/genetics , beta Catenin/metabolism , gamma Catenin/genetics
2.
Nat Commun ; 8(1): 593, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28928446

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are involved in human diseases, such as allergy, atopic dermatitis and nasal polyposis, but their function in human cancer remains unclear. Here we show that, in acute promyelocytic leukaemia (APL), ILC2s are increased and hyper-activated through the interaction of CRTH2 and NKp30 with elevated tumour-derived PGD2 and B7H6, respectively. ILC2s, in turn, activate monocytic myeloid-derived suppressor cells (M-MDSCs) via IL-13 secretion. Upon treating APL with all-trans retinoic acid and achieving complete remission, the levels of PGD2, NKp30, ILC2s, IL-13 and M-MDSCs are restored. Similarly, disruption of this tumour immunosuppressive axis by specifically blocking PGD2, IL-13 and NKp30 partially restores ILC2 and M-MDSC levels and results in increased survival. Thus, using APL as a model, we uncover a tolerogenic pathway that may represent a relevant immunosuppressive, therapeutic targetable, mechanism operating in various human tumour types, as supported by our observations in prostate cancer.Group 2 innate lymphoid cells (ILC2s) modulate inflammatory and allergic responses, but their function in cancer immunity is still unclear. Here the authors show that, in acute promyelocytic leukaemia, tumour-activated ILC2s secrete IL-13 to induce myeloid-derived suppressor cells and support tumour growth.


Subject(s)
B7 Antigens/immunology , Lymphocytes/immunology , Myeloid-Derived Suppressor Cells/immunology , Natural Cytotoxicity Triggering Receptor 3/immunology , Prostaglandin D2/immunology , A549 Cells , Animals , Antineoplastic Agents/therapeutic use , B7 Antigens/metabolism , Cell Line, Tumor , Disease Models, Animal , HL-60 Cells , Hep G2 Cells , Humans , Immunity, Innate/immunology , Interleukin-13/immunology , Interleukin-13/metabolism , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/metabolism , Lymphocytes/metabolism , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/metabolism , Natural Cytotoxicity Triggering Receptor 3/metabolism , Prostaglandin D2/metabolism , Protein Binding , Tretinoin/therapeutic use
5.
ACS Chem Biol ; 11(5): 1422-7, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26929989

ABSTRACT

Short α-helical peptides stabilized by linkages between constituent amino acids offer an attractive format for ligand development. In recent years, a range of excellent ligands based on stabilized α-helices were generated by rational design using α-helical peptides of natural proteins as templates. Herein, we developed a method to engineer chemically stabilized α-helical ligands in a combinatorial fashion. In brief, peptides containing cysteines in position i and i + 4 are genetically encoded by phage display, the cysteines are modified with chemical bridges to impose α-helical conformations, and binders are isolated by affinity selection. We applied the strategy to affinity mature an α-helical peptide binding ß-catenin. We succeeded in developing ligands with Kd's as low as 5.2 nM, having >200-fold improved affinity. The strategy is generally applicable for affinity maturation of any α-helical peptide. Compared to hydrocarbon stapled peptides, the herein evolved thioether-bridged peptide ligands can be synthesized more easily, as no unnatural amino acids are required and the cyclization reaction is more efficient and yields no stereoisomers. A further advantage of the thioether-bridged peptide ligands is that they can be expressed recombinantly as fusion proteins.


Subject(s)
Bacteriophages/metabolism , Ligands , Amino Acid Sequence , Molecular Conformation , Molecular Sequence Data , Peptide Library , Peptides/chemistry , beta Catenin/metabolism
6.
Nat Commun ; 6: 10004, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26643973

ABSTRACT

Despite the recent evidence of the existence of myelodysplastic syndrome (MDS) stem cells in 5q-MDS patients, it is unclear whether haematopoietic stem cells (HSCs) could also be the initiating cells in other MDS subgroups. Here we demonstrate that SF3B1 mutation(s) in our cohort of MDS patients with ring sideroblasts can arise from CD34(+)CD38(-)CD45RA(-)CD90(+)CD49f(+) HSCs and is an initiating event in disease pathogenesis. Xenotransplantation of SF3B1 mutant HSCs leads to persistent long-term engraftment restricted to myeloid lineage. Moreover, genetically diverse evolving subclones of mutant SF3B1 exist in mice, indicating a branching multi-clonal as well as ancestral evolutionary paradigm. Subclonal evolution in mice is also seen in the clinical evolution in patients. Sequential sample analysis shows clonal evolution and selection of the malignant driving clone leading to AML transformation. In conclusion, our data show SF3B1 mutations can propagate from HSCs to myeloid progeny, therefore providing a therapeutic target.


Subject(s)
Bone Marrow/metabolism , Cell Transformation, Neoplastic/genetics , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Aged , Animals , Female , Genotype , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Male , Mice , Middle Aged , Mutation , Neoplasm Transplantation , RNA Splicing Factors , Young Adult
7.
Stem Cells Transl Med ; 3(4): 520-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24493855

ABSTRACT

Acute myeloid leukemia-initiating cells (LICs) are responsible for the emergence of leukemia and relapse after chemotherapy. Despite their identification more than 15 years ago, our understanding of the mechanisms responsible for their self-renewal activity and their chemoresistance remains poor. The slow progress in this area is partly due to the difficulty of studying these cells ex vivo. Indeed, current studies are reliant on xenotransplantation assays in immunodeficient mice. In this paper, we report that by modeling key elements of the bone marrow niche using different stromal feeder layers and hypoxic culture conditions, we can maintain LICs over at least 3 weeks and support their self-renewal properties demonstrated through primary and secondary successful xenograft. We provide a proof of principle that this niche-like culture system can be used to study LIC chemoresistance following in vitro cytarabine treatment similarly to the xenograft chemotherapy model. We found that although LICs are believed to be more chemoresistant than non-LICs, functionally defined LICs are not enriched after cytarabine treatment, and heterogeneity in their resistance to treatment can be seen between patients and even within the same patient. We present a culture system that can be used as an in vitro surrogate for xenotransplantation and that has the potential to dramatically increase the throughput of the investigation of LICs. This would further provide the means by which to identify and target the functionality of the different signaling pathways involved in the maintenance and resistance of LICs to improve acute myeloid leukemia treatments.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/metabolism , Stem Cell Niche , Animals , Female , Heterografts , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/transplantation , Signal Transduction/drug effects , Tumor Cells, Cultured
8.
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
9.
Br J Haematol ; 161(3): 389-401, 2013 May.
Article in English | MEDLINE | ID: mdl-23432359

ABSTRACT

Current therapeutic regimens for acute myeloid leukaemia (AML) are still associated with high rates of relapse. Immunotherapy with T-cells genetically modified to express chimeric antigen receptors (CARs) represents an innovative approach. Here we investigated the targeting of the interleukin three receptor alpha (IL3RA; CD123) molecule, which is overexpressed on AML bulk population, CD34(+) leukaemia progenitors, and leukaemia stem cells (LSC) compared to normal haematopoietic stem/progenitor cells (HSPCs), and whose overexpression is associated with poor prognosis. Cytokine-induced killer (CIK) cells were transduced with SFG-retroviral-vector encoding an anti-CD123 CAR. Transduced cells were able to strongly kill CD123(+) cell lines, as well as primary AML blasts. Interestingly, secondary colony experiments demonstrated that anti-CD123.CAR preserved in vitro HSPCs, in contrast to a previously generated anti-CD33.CAR, while keeping an identical cytotoxicity profile towards AML. Furthermore, limited killing of normal monocytes and CD123-low-expressing endothelial cells was noted, thus indicating a low toxicity profile of the anti-CD123.CAR. Taken together, our results indicate that CD123-specific CARs strongly enhance anti-AML CIK functions, while sparing HSPCs and normal low-expressing antigen cells, paving the way to develop novel immunotherapy approaches for AML treatment.


Subject(s)
Cytokine-Induced Killer Cells/immunology , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/pathology , Receptors, Cell Surface/physiology , Cell Line, Tumor/metabolism , Coculture Techniques , Cytokines/metabolism , Cytotoxicity Tests, Immunologic , Endothelial Cells , Female , HEK293 Cells , Hematopoietic Stem Cells , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Monocytic, Acute/pathology , Male , Monocytes , Recombinant Fusion Proteins/physiology , Transduction, Genetic , Tumor Stem Cell Assay
10.
Hum Gene Ther Methods ; 23(6): 376-86, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23186165

ABSTRACT

Use of adoptive T-cell therapy (ACT) is increasing; however, T-cell therapy can result in severe toxicity. Consequently, several suicide-gene strategies that allow selective destruction of the infused T cells have been described. We compared effectiveness of four such strategies in vitro in Epstein Barr virus (EBV)-cytotoxic T lymphocytes (CTLs). Herpes simplex virus thymidine kinase (HSV-TK), human inducible caspase 9 (iCasp9), mutant human thymidylate kinase (mTMPK), and human CD20 codon optimized genes were cloned in frame with 2A-truncated codon optimized CD34 (dCD34) in a retroviral vector. Codon-optimization considerably improved CD20 expression. EBV-CTLs could be efficiently transduced in all constructs, with transgene expression similar to the control vector containing dCD34 alone. Expression was maintained for prolonged cultures. Expression of the suicide genes was not associated with alterations in immunophenotype, proliferation, or function of CTLs. Activation of HSV-TK, iCasp9, and CD20 ultimately resulted in equally effective destruction of transduced T cells. However, while iCasp9 and CD20 effected immediate cell-death induction, HSV-TK-expressing T cells required 3 days of exposure to ganciclovir to reach full effect. mTMPK-transduced cells showed lower T-cell killing all time points. Our results suggest that the faster activity of iCasp9 might be advantageous in treating certain types of acutely life-threatening toxicity. Codon-optimized CD20 has potential as a suicide gene.


Subject(s)
Caspase 9/genetics , Genes, Transgenic, Suicide/genetics , T-Lymphocytes, Cytotoxic/metabolism , Thymidine Kinase/genetics , Antigens, CD20/genetics , Antigens, CD34/genetics , Ganciclovir/pharmacology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mutation , Simplexvirus/enzymology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
11.
Ital J Pediatr ; 37: 46, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21939556

ABSTRACT

Leukaemia immunotherapy represents a fascinating and promising field of translational research, particularly as an integrative approach of bone marrow transplantation. Adoptive immunotherapy by the use of donor-derived expanded leukaemia-specific T cells has showed some kind of clinical response, but the major advance is nowadays represented by gene manipulation of donor immune cells, so that they acquire strict specificity towards the tumour target and potent lytic activity, followed by significant proliferation, increased survival and possibly anti-tumour memory state. This is achieved by gene insertion of Chimeric T-cell Antigen Receptors (CARs), which are artificial molecules containing antibody-derived fragments (to bind the specific target), joined with potent signalling T-Cell Receptor (TCR)-derived domains that activate the manipulated cells. This review will discuss the main application of this approach particularly focusing on the paediatric setting, raising advantages and disadvantages and discussing relevant perspectives of use in the nearest future.


Subject(s)
Immunotherapy, Adoptive/trends , Leukemia/therapy , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/immunology , Humans , Immunotherapy, Adoptive/methods , Leukemia, Lymphoid/therapy , Leukemia, Myeloid/therapy , Peptide Fragments/immunology , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , Signal Transduction
12.
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
13.
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
14.
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
15.
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
16.
Circulation ; 120(8): 699-708, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19667236

ABSTRACT

BACKGROUND: Immune responses participate in several phases of atherosclerosis; there is, in fact, increasing evidence that both adaptive immunity and innate immunity tightly regulate atherogenesis. Pentraxins are a superfamily of acute-phase proteins that includes short pentraxins such as C-reactive protein or long pentraxins such as PTX3, a molecule acting as the humoral arm of innate immunity. To address the potential role of PTX3 in atherogenesis, we first investigated the expression of PTX3 during atherogenesis, generated double-knockout mice lacking PTX3 and apolipoprotein E, and then studied the effect of murine PTX3 deficiency on plasma lipids, atherosclerosis development, and gene expression pattern in the vascular wall. METHODS AND RESULTS: PTX3 expression increases in the vascular wall of apolipoprotein E-knockout mice from 3 up to 18 months of age. Double-knockout mice lacking PTX3 and apolipoprotein E were fed an atherogenic diet for 16 weeks. Aortic lesions were significantly increased in double-knockout mice and mice heterozygous for PTX3 compared with apolipoprotein E-knockout mice. Mice lacking PTX3 showed a more pronounced inflammatory profile in the vascular wall as detected by cDNA microarray and quantitative polymerase chain reaction analysis and an increased macrophage accumulation within the plaque. Finally, lesion size correlated with the number of bone marrow monocytes. CONCLUSIONS: PTX3 has atheroprotective effects in mice, which, in light of the cardioprotective effects recently reported, suggests a cardiovascular protective function of the long pentraxin 3 through the modulation of the immunoinflammatory balance in the cardiovascular system.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/immunology , C-Reactive Protein/genetics , C-Reactive Protein/immunology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Vasculitis/genetics , Vasculitis/immunology , Animals , Aorta/pathology , Aorta/physiology , Apolipoproteins E/genetics , Atherosclerosis/pathology , Body Weight , Female , Gene Expression/immunology , Lipids/blood , Macrophages/pathology , Male , Mice , Mice, Knockout , Sex Factors , Vasculitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...