45 results on '"Pizzitola I"'
Search Results
2. Chimeric antigen receptor: a cell therapy based approach for the treatment of acute myeloid leukemia
- Author
-
BONNET, DOMINIQUE, Pizzitola, I, BIAGI, ETTORE, PIZZITOLA, IRENE, BONNET, DOMINIQUE, Pizzitola, I, BIAGI, ETTORE, and PIZZITOLA, IRENE
- Abstract
Despite the progress in the treatment of acute myeloid leukemia (AML) achieved in the last decades, a significant number of patients are still refractory to or relapse after standard cures. Hence, to improve cure rates of AML, it is crucial to develop novel therapeutic strategies. Immunotherapy with T cells genetically modified to express chimeric antigen receptors (CARs) represent a valid option in this sense. CARs are artificial molecules constituted by an extracellular-antigen-binding domain derived from a monoclonal antibody and an intracellular-signalling region that is immediately triggered after antigen recognition. Therefore, CARs combine the antigen binding properties of mononoclonal antibodies to T cell mediated effector functions, including the killing mechanism -that might be active against antibody resistant targets-, cytokine secretion- that might boost the anti-tumoral immune response- and capacity to efficiently home and infiltrate tumor sites. Different CARs have been generated so far, against a wide range of surface molecules expressed by many tumors and, currently, several phase I clinical trials are undergoing and the results obtained so far are very encouraging. The CARs approach can be employed to selectively target AML cells due to the overexpression of myeloid antigens, like CD33 and CD123. We recently demonstrated that expression of CD33-specific CARs in a population of ex-vivo activated T cells, called “cytokine induced killer” (CIK) cells, confers them potent in vitro anti-leukemic functions. However, since CD33 antigen is also expressed on normal haematopoietic stem/progenitors cells (HSPCs) resulting in a potential severe impairment of normal myelopoiesis, CD123 has recently been proposed as a new potential attractive molecule based on its differential expression pattern, being widely overexpressed by AML population and at the same time less expressed on HSPCs. In order to improve the safety profile against these cells we develop and tes
- Published
- 2013
3. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor
- Author
-
Tettamanti, S, Marin, V, Pizzitola, I, Magnani, C, GIORDANO ATTIANESE, G, Cribioli, E, Maltese, F, Galimberti, S, Lopez, A, Biondi, A, Bonnet, D, Biagi, E, PIZZITOLA, IRENE, MAGNANI, CHIARA FRANCESCA, GIORDANO ATTIANESE, GRETA MARIA PAOLA, GALIMBERTI, STEFANIA, BIONDI, ANDREA, BIAGI, ETTORE, Tettamanti, S, Marin, V, Pizzitola, I, Magnani, C, GIORDANO ATTIANESE, G, Cribioli, E, Maltese, F, Galimberti, S, Lopez, A, Biondi, A, Bonnet, D, Biagi, E, PIZZITOLA, IRENE, MAGNANI, CHIARA FRANCESCA, GIORDANO ATTIANESE, GRETA MARIA PAOLA, GALIMBERTI, STEFANIA, BIONDI, ANDREA, and BIAGI, ETTORE
- 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.
- Published
- 2013
4. Chimeric Antigen Receptor for Specific Targeting of Acute Myeloid Leukemia
- Author
-
Pizzitola, I, Anjos Afonso, F, Rouault Pierre, K, Lassailly, F, Tettamanti, S, Biondi, A, Biagi, E, Bonnet, D, PIZZITOLA, IRENE, BIONDI, ANDREA, BIAGI, ETTORE, Bonnet, D., Pizzitola, I, Anjos Afonso, F, Rouault Pierre, K, Lassailly, F, Tettamanti, S, Biondi, A, Biagi, E, Bonnet, D, PIZZITOLA, IRENE, BIONDI, ANDREA, BIAGI, ETTORE, and Bonnet, D.
- Published
- 2012
5. Targeting of Acute Myeloid Leukemia by Cytokine-Induced Killer Cells Redirected with a Novel CD123-Specific Chimeric Antigen Receptor
- Author
-
Tettamanti, S, Marin, V, Pizzitola, I, Magnani, C, Attianese, G, Cribioli, E, Lopez, A, Biondi, A, Bonnet, D, Biagi, E, PIZZITOLA, IRENE, MAGNANI, CHIARA FRANCESCA, BIONDI, ANDREA, BIAGI, ETTORE, Tettamanti, S, Marin, V, Pizzitola, I, Magnani, C, Attianese, G, Cribioli, E, Lopez, A, Biondi, A, Bonnet, D, Biagi, E, PIZZITOLA, IRENE, MAGNANI, CHIARA FRANCESCA, BIONDI, ANDREA, and BIAGI, ETTORE
- Published
- 2012
6. Comparison of different suicide-gene strategies for the safety improvement of genetically manipulated T cells
- Author
-
Marin, V, Cribioli, E, Philip, B, Tettamanti, S, Pizzitola, I, Biondi, A, Biagi, E, Pule, M, PIZZITOLA, IRENE, BIONDI, ANDREA, BIAGI, ETTORE, Pule, M., Marin, V, Cribioli, E, Philip, B, Tettamanti, S, Pizzitola, I, Biondi, A, Biagi, E, Pule, M, PIZZITOLA, IRENE, BIONDI, ANDREA, BIAGI, ETTORE, and Pule, M.
- 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
- Published
- 2012
7. In Vitro and In Vivo Antitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL against CD33 Acute Myeloid Leukemia
- Author
-
Dutour, A, Marin, V, Pizzitola, I, Valsesia Wittmann, S, Lee, D, Yvon, E, Finney, H, Lawson, A, Brenner, M, Biondi, A, Biagi, E, Rousseau, R, BIONDI, ANDREA, BIAGI, ETTORE, Rousseau, R., Dutour, A, Marin, V, Pizzitola, I, Valsesia Wittmann, S, Lee, D, Yvon, E, Finney, H, Lawson, A, Brenner, M, Biondi, A, Biagi, E, Rousseau, R, BIONDI, ANDREA, BIAGI, ETTORE, and Rousseau, R.
- Abstract
Genetic engineering of T cells with chimeric T-cell receptors (CARs) is an attractive strategy to treat malignancies. It extends the range of antigens for adoptive T-cell immunotherapy, and major mechanisms of tumor escape are bypassed. With this strategy we redirected immune responses towards the CD33 antigen to target acute myeloid leukemia. To improve in vivo T-cell persistence, we modified human Epstein Barr Virus-(EBV-) specific cytotoxic T cells with an anti-CD33.CAR. Genetically modified T cells displayed EBV and HLA-unrestricted CD33 bispecificity in vitro. In addition, though showing a myeloablative activity, they did not irreversibly impair the clonogenic potential of normal CD34(+) hematopoietic progenitors. Moreover, after intravenous administration into CD33(+) human acute myeloid leukemia-bearing NOD-SCID mice, anti-CD33-EBV-specific T cells reached the tumor sites exerting antitumor activity in vivo. In conclusion, targeting CD33 by CAR-modified EBV-specific T cells may provide additional therapeutic benefit to AML patients as compared to conventional chemotherapy or transplantation regimens alone.
- Published
- 2012
8. New advances in leukaemia immunotherapy by the use of Chimeric Artificial Antigen Receptors (CARs): state of the art and perspectives for the near future
- Author
-
Biagi, E, Marin, V, Giordano Attianese, G, Pizzitola, I, Tettamanti, S, Cribioli, E, Biondi, A, BIAGI, ETTORE, BIONDI, ANDREA, Giordano Attianese, GMP, Biagi, E, Marin, V, Giordano Attianese, G, Pizzitola, I, Tettamanti, S, Cribioli, E, Biondi, A, BIAGI, ETTORE, BIONDI, ANDREA, and Giordano Attianese, GMP
- 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 applications of this approach particularly focusing on the paediatric setting, raising advantages and disadvantages and discussing relevant perspectives of use in the nearest future.
- Published
- 2011
9. Exploration of the lysis mechanisms of leukaemic blasts by chimaeric T-cells
- Author
-
Laurin, D, Marin, V, Biagi, E, Pizzitola, I, Agostoni, V, Gallot, G, Vié, H, Jacob, M, Chaperot, L, Aspord, C, Plumas, J, BIAGI, ETTORE, PIZZITOLA, IRENE, Jacob, MC, Plumas, J., Laurin, D, Marin, V, Biagi, E, Pizzitola, I, Agostoni, V, Gallot, G, Vié, H, Jacob, M, Chaperot, L, Aspord, C, Plumas, J, BIAGI, ETTORE, PIZZITOLA, IRENE, Jacob, MC, and Plumas, J.
- 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- and TNF- 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. Copyright © 2010 David Laurin et al.
- Published
- 2010
10. Cytokine Induced Killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors
- Author
-
Marin, V, Pizzitola, I, Agostoni, V, Giordano Attianese, G, Finney, H, Lawson, A, Pule, M, Rousseau, R, Biondi, A, Biagi, E, PIZZITOLA, IRENE, Giordano Attianese, GMP, BIONDI, ANDREA, BIAGI, ETTORE, Marin, V, Pizzitola, I, Agostoni, V, Giordano Attianese, G, Finney, H, Lawson, A, Pule, M, Rousseau, R, Biondi, A, Biagi, E, PIZZITOLA, IRENE, Giordano Attianese, GMP, BIONDI, ANDREA, and BIAGI, ETTORE
- Abstract
Background. Cytokine induced killer cells are ex-vivo expanded cells with potent antitumoral activity. Cytokine induced killer cells infusion in acute myeloid leukemia patients relapsing after allogeneic hematopoietic stem cell transplant is well tolerated, but limited clinical responses were 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-zeta and anti-CD33-CD28-OX40-zeta chimeric receptors were used to transduce cytokine induced killer cells. Transduced cells were in vitro characterized for their ability to lyse leukemic targets (4-hour-51Chromium-release and 6-day co-cultures assays on human stromal mesenchymal cells), to proliferate (3H-thymidine-incorporation assay) and to secrete cytokines (Flow cytomix assay) after contact with acute myeloid leukemia cells. Their activity against normal CD34+ hematopoietic progenitors 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 a prominent CD33-specific proliferative activity, with a release of high levels of immunostimulatory cytokines. The presence of CD28-OX40 in chimeric receptor endodomain was associated with a significant amelioration of cytokine induced killer cells anti-leukemic activity. Importantly, even though anti-CD33 chimeric receptors-transduced cytokine induced killer cells showed toxicity against normal hematopoietic CD34+ progenitors, a residual clonogenic activ
- Published
- 2010
11. Chimeric antigen receptor for specific targeting of acute myeloid leukemia
- Author
-
Pizzitola, I., primary, Anjos-Afonso, F., additional, Rouault-Pierre, K., additional, Lassailly, F., additional, Biondi, A., additional, Biagi, E., additional, and Bonnet, D., additional
- Published
- 2013
- Full Text
- View/download PDF
12. Targeting of acute myeloid leukemia (AML) by cytokine-induced killer (CIK) cells redirected with a novel CD123-specific chimeric antigen receptor (CAR)
- Author
-
Tettamanti, S., primary, Marin, V., additional, Pizzitola, I., additional, Magnani, C.F., additional, Giordano Attianese, G.M., additional, Cribioli, E., additional, Maltese, F., additional, Lopez, A., additional, Biondi, A., additional, Bonnet, D., additional, and Biagi, E., additional
- Published
- 2013
- Full Text
- View/download PDF
13. In VitroandIn VivoAntitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL againstCD33+Acute Myeloid Leukemia
- Author
-
Dutour, A., primary, Marin, V., additional, Pizzitola, I., additional, Valsesia-Wittmann, S., additional, Lee, D., additional, Yvon, E., additional, Finney, H., additional, Lawson, A., additional, Brenner, M., additional, Biondi, A., additional, Biagi, E., additional, and Rousseau, R., additional
- Published
- 2012
- Full Text
- View/download PDF
14. Cytokine-induced killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors
- Author
-
Marin, V., primary, Pizzitola, I., additional, Agostoni, V., additional, Attianese, G. M. P. G., additional, Finney, H., additional, Lawson, A., additional, Pule, M., additional, Rousseau, R., additional, Biondi, A., additional, and Biagi, E., additional
- Published
- 2010
- Full Text
- View/download PDF
15. 300 A novel immunotherapy approach for B-CLL by use of Chimeric TCR
- Author
-
Giordano Attianese, G., primary, Hoyos, V., additional, Marin, V., additional, Savoldo, B., additional, Pizzitola, I., additional, Agostoni, V., additional, Parma, M., additional, Biondi, A., additional, Dotti, G., additional, and Biagi, E., additional
- Published
- 2010
- Full Text
- View/download PDF
16. In Vitro and In Vivo Antitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL against CD33+ Acute Myeloid Leukemia.
- Author
-
Dutour, A., Marin, V., Pizzitola, I., Valsesia-Wittmann, S., Lee, D., Yvon, E., Finney, H., Lawson, A., Brenner, M., Biondi, A., Biagi, E., and Rousseau, R.
- Subjects
ACUTE myeloid leukemia ,ANTINEOPLASTIC agents ,EPSTEIN-Barr virus ,IMMUNOTHERAPY ,IMMUNE response ,CHIMERIC proteins ,T-cell receptor genes ,GENETIC engineering - Abstract
Genetic engineering of T cells with chimeric T-cell receptors (CARs) is an attractive strategy to treat malignancies. It extends the range of antigens for adoptive T-cell immunotherapy, and major mechanisms of tumor escape are bypassed. With this strategy we redirected immune responses towards the CD33 antigen to target acute myeloid leukemia. To improve in vivo T-cell persistence, we modified human Epstein Barr Virus-(EBV-) specific cytotoxic T cells with an anti-CD33.CAR. Genetically modified T cells displayed EBV and HLA-unrestricted CD33 bispecificity in vitro. In addition, though showing a myeloablative activity, they did not irreversibly impair the clonogenic potential of normal CD34
+ hematopoietic progenitors. Moreover, after intravenous administration into CD33+ human acutemyeloid leukemia-bearing NOD-SCID mice, anti-CD33-EBV-specific T cells reached the tumor sites exerting antitumor activity in vivo. In conclusion, targeting CD33 by CAR-modified EBV-specific T cells may provide additional therapeutic benefit to AML patients as compared to conventional chemotherapy or transplantation regimens alone. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
17. New advances in leukaemia immunotherapy by the use of Chimeric Artificial Antigen Receptors (CARs): state of the art and perspectives for the near future
- Author
-
Cribioli Elisabetta, Tettamanti Sarah, Pizzitola Irene, Attianese Greta, Marin Virna, Biagi Ettore, and Biondi Andrea
- Subjects
Leukaemia immunotherapy ,cell therapy ,gene therapy ,chimeric artificial receptors ,Pediatrics ,RJ1-570 - Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
18. γ-Catenin-Dependent Signals Maintain BCR-ABL1+ B Cell Acute Lymphoblastic Leukemia
- Author
-
Jean-Pierre Bourquin, Beat Bornhauser, Beena Jeevan-Raj, Oliver Hantschel, Werner Held, Joerg Huelsken, Maxime Danilo, Margot Thome, Georgi S Angelov, Anja Irmisch, Noemie Luong-Gardiol, Gianni Cazzaniga, Sara Curtet, Mélanie Charmoy, Yuna Huang, Yves Chalandon, Irene Pizzitola, Imran Siddiqui, Mélanie Juilland, University of Zurich, Held, Werner, Luong-Gardiol, N, Siddiqui, I, Pizzitola, I, Jeevan-Raj, B, Charmoy, M, Huang, Y, Irmisch, A, Curtet, S, Angelov, G, Danilo, M, Juilland, M, Bornhauser, B, Thome, M, Hantschel, O, Chalandon, Y, Cazzaniga, G, Bourquin, J, Huelsken, J, and Held, W
- Subjects
0301 basic medicine ,Cancer Research ,Survivin ,Fusion Proteins, bcr-abl ,MYC ,Mice, SCID ,1307 Cell Biology ,0302 clinical medicine ,Animals ,Fusion Proteins, bcr-abl/genetics ,Fusion Proteins, bcr-abl/metabolism ,Gene Expression Regulation, Leukemic ,Humans ,K562 Cells ,Mice, Inbred NOD ,Mice, Transgenic ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology ,Proto-Oncogene Proteins c-myc/genetics ,Proto-Oncogene Proteins c-myc/metabolism ,Survivin/genetics ,Survivin/metabolism ,Wnt Signaling Pathway ,beta Catenin/genetics ,beta Catenin/metabolism ,gamma Catenin/genetics ,gamma Catenin/metabolism ,B cell acute lymphoblastic leukemia (B-ALL) ,BCR-ABL1 ,BIRC5 (Survivin) ,chronic myeloid leukemia (CML) ,junction plakoglobin ,β-catenin ,γ-catenin ,hemic and lymphatic diseases ,Chronic myeloid leukemia (CML) ,1306 Cancer Research ,beta Catenin ,ddc:616 ,biology ,Wnt signaling pathway ,Myeloid leukemia ,3. Good health ,Oncology ,030220 oncology & carcinogenesis ,2730 Oncology ,Junction plakoglobin ,Β-catenin ,Human ,Beta-catenin ,Plakoglobin ,610 Medicine & health ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,K562 Cell ,Animal ,Cell Biology ,Fusion protein ,030104 developmental biology ,10036 Medical Clinic ,Catenin ,biology.protein ,Cancer research ,gamma Catenin ,K562 cells - 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.
- Published
- 2019
- Full Text
- View/download PDF
19. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo
- Author
-
Orietta Spinelli, Irene Pizzitola, Francois Lassailly, Fernando Anjos-Afonso, Sarah Tettamanti, Andrea Biondi, Dominique Bonnet, Kevin Rouault-Pierre, Ettore Biagi, Pizzitola, I, Anjos Afonso, F, Rouault Pierre, K, Lassailly, F, Tettamanti, S, Spinelli, O, Biondi, A, Biagi, E, and Bonnet, D
- Subjects
Cancer Research ,Myeloid ,Recombinant Fusion Proteins ,T-Lymphocytes ,Sialic Acid Binding Ig-like Lectin 3 ,CD33 ,Interleukin-3 Receptor alpha Subunit ,Mice, SCID ,Biology ,Mice ,Cytokine-Induced Killer Cells ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Progenitor cell ,Myeloid leukemia ,Hematology ,medicine.disease ,Chimeric antigen receptor ,Leukemia, Myeloid, Acute ,Receptors, Antigen ,AML, CD123, CD33, chimeric antigen receptor ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,Oncology ,Immunology ,Stem cell - Abstract
As significant numbers of acute myeloid leukemia (AML) patients are still refractory to conventional therapies or experience relapse, immunotherapy using T-cells expressing chimeric antigen receptors (CARs) might represent a valid treatment option. AML cells frequently overexpress the myeloid antigens CD33 and CD123, for which specific CARs can be generated. However, CD33 is also expressed on normal hematopoietic stem/progenitors cells (HSPCs), and its targeting could potentially impair normal hematopoiesis. In contrast, CD123 is widely expressed by AML, while low expression is detected on HSPCs, making it a much more attractive target. In this study we describe the in vivo efficacy and safety of using cytokine-induced-killer (CIK) cells genetically modified to express anti-CD33 or anti-CD123 CAR to target AML. We show that both these modified T-cells are very efficient in reducing leukemia burden in vivo, but only the anti-CD123 CAR has limited killing on normal HSPCs, thus making it a very attractive immunotherapeutic tool for AML treatment.Leukemia accepted article preview online, 7 February 2014; doi:10.1038/leu.2014.62.
- Published
- 2014
- Full Text
- View/download PDF
20. Comparison of Different Suicide-Gene Strategies for the Safety Improvement of Genetically Manipulated T Cells
- Author
-
Virna Marin, Sarah Tettamanti, Ettore Biagi, Irene Pizzitola, Martin Pule, Andrea Biondi, Elisabetta Cribioli, Brian Philip, Marin, V, Cribioli, E, Philip, B, Tettamanti, S, Pizzitola, I, Biondi, A, Biagi, E, and Pule, M
- Subjects
Transgene ,Genetic enhancement ,Genetic Vectors ,Antigens, CD34 ,Biology ,medicine.disease_cause ,Thymidine Kinase ,Applied Microbiology and Biotechnology ,Thymidylate kinase ,Viral vector ,Interferon-gamma ,Antigen ,Genetics ,medicine ,Humans ,Simplexvirus ,Ganciclovir ,Research Articles ,Genetics (clinical) ,Pharmacology ,Genes, Transgenic, Suicide ,Suicide gene ,Antigens, CD20 ,Virology ,Epstein–Barr virus ,Caspase 9 ,gene therapy, suicide gene, iCasp9, HSV-Tk ,Killer Cells, Natural ,Thymidine kinase ,Mutation ,Cancer research ,Molecular Medicine ,T-Lymphocytes, Cytotoxic - 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.
- Published
- 2012
- Full Text
- View/download PDF
21. In vitro and in vivo model of a novel immunotherapy approach for chronic lymphocytic leukemia by anti-CD23 chimeric antigen receptor
- Author
-
Maurilio Ponzoni, Valentina Agostoni, Virna Marin, Sarah Tettamanti, Irene Pizzitola, Paolo Ghia, Ettore Biagi, Matteo Parma, Greta Maria Paola Giordano Attianese, Valentina Hoyos, Gianpietro Dotti, Barbara Savoldo, Andrea Biondi, Maria Teresa Sabrina Bertilaccio, Giordano Attianese, G, Marin, V, Hoyos, V, Savoldo, B, Pizzitola, I, Tettamanti, S, Agostoni, V, Parma, M, Ponzoni, M, Bertilaccio, M, Ghia, P, Biondi, A, Dotti, G, Biagi, E, Giordano Attianese, Gmp, Ponzoni, Maurilio, Bertilaccio, Mt, Ghia, PAOLO PROSPERO, and Biagi, E.
- Subjects
Cytotoxicity, Immunologic ,Cell therapy, CLL, chimeric TCR, CD23 ,Recombinant Fusion Proteins ,T-Lymphocytes ,Immunology ,Gene Expression ,Biology ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Biochemistry ,Mice ,Interleukin 21 ,stomatognathic system ,Antigen ,immune system diseases ,Cell Line, Tumor ,hemic and lymphatic diseases ,Animals ,Humans ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Mice, Knockout ,B-Lymphocytes ,Receptors, IgE ,ZAP70 ,hemic and immune systems ,Gene Therapy ,Cell Biology ,Hematology ,Natural killer T cell ,Leukemia, Lymphocytic, Chronic, B-Cell ,Xenograft Model Antitumor Assays ,Molecular biology ,Coculture Techniques ,DNA-Binding Proteins ,Cytokines ,Interleukin-2 ,CD5 - 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-beta, 20-fold more TNF-alpha, and 4-fold more IFN-gamma). 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(-/-)gamma(-/-)(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. (Blood. 2011; 117(18): 4736-4745) 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-beta, 20-fold more TNF-alpha, and 4-fold more IFN-gamma). 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(-/-)gamma(-/-)(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. (Blood. 2011; 117(18): 4736-4745)
- Published
- 2011
- Full Text
- View/download PDF
22. Exploration of the Lysis Mechanisms of Leukaemic Blasts by Chimaeric T-Cells
- Author
-
Laurence Chaperot, Virna Marin, Valentina Agostoni, Joel Plumas, Henri Vié, Caroline Aspord, David Laurin, Ettore Biagi, Géraldine Gallot, Marie Christine Jacob, Irene Pizzitola, Laurin, D, Marin, V, Biagi, E, Pizzitola, I, Agostoni, V, Gallot, G, Vié, H, Jacob, M, Chaperot, L, Aspord, C, and Plumas, J
- Subjects
Adoptive cell transfer ,Article Subject ,lcsh:Biotechnology ,Health, Toxicology and Mutagenesis ,Antigens, CD19 ,lcsh:Medicine ,Apoptosis ,chemical and pharmacologic phenomena ,lcsh:Chemical technology ,Immunotherapy, Adoptive ,lcsh:Technology ,Granzymes ,Cell Line ,Cytokine-Induced Killer Cells ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,lcsh:TP248.13-248.65 ,Genetics ,Humans ,Cytotoxic T cell ,lcsh:TP1-1185 ,Molecular Biology ,biology ,Cytokine-induced killer cell ,Perforin ,lcsh:T ,lcsh:R ,Degranulation ,Lysosome-Associated Membrane Glycoproteins ,hemic and immune systems ,Receptors, Death Domain ,General Medicine ,Flow Cytometry ,Cytolysis ,CTL ,Cell Transformation, Neoplastic ,Granzyme ,CTL, CIK, immunotherapy ,biology.protein ,Cancer research ,Molecular Medicine ,Research Article ,Signal Transduction ,T-Lymphocytes, Cytotoxic ,Biotechnology - 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- and TNF- 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. Copyright © 2010 David Laurin et al.
- Published
- 2010
- Full Text
- View/download PDF
23. Chimeric antigen receptor: a cell therapy based approach for the treatment of acute myeloid leukemia
- Author
-
PIZZITOLA, IRENE, Pizzitola, I, and BIAGI, ETTORE
- Subjects
MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,acute Myeloid Leukemia, immunotherapy, chimeric antigen receptor - Abstract
Despite the progress in the treatment of acute myeloid leukemia (AML) achieved in the last decades, a significant number of patients are still refractory to or relapse after standard cures. Hence, to improve cure rates of AML, it is crucial to develop novel therapeutic strategies. Immunotherapy with T cells genetically modified to express chimeric antigen receptors (CARs) represent a valid option in this sense. CARs are artificial molecules constituted by an extracellular-antigen-binding domain derived from a monoclonal antibody and an intracellular-signalling region that is immediately triggered after antigen recognition. Therefore, CARs combine the antigen binding properties of mononoclonal antibodies to T cell mediated effector functions, including the killing mechanism -that might be active against antibody resistant targets-, cytokine secretion- that might boost the anti-tumoral immune response- and capacity to efficiently home and infiltrate tumor sites. Different CARs have been generated so far, against a wide range of surface molecules expressed by many tumors and, currently, several phase I clinical trials are undergoing and the results obtained so far are very encouraging. The CARs approach can be employed to selectively target AML cells due to the overexpression of myeloid antigens, like CD33 and CD123. We recently demonstrated that expression of CD33-specific CARs in a population of ex-vivo activated T cells, called “cytokine induced killer” (CIK) cells, confers them potent in vitro anti-leukemic functions. However, since CD33 antigen is also expressed on normal haematopoietic stem/progenitors cells (HSPCs) resulting in a potential severe impairment of normal myelopoiesis, CD123 has recently been proposed as a new potential attractive molecule based on its differential expression pattern, being widely overexpressed by AML population and at the same time less expressed on HSPCs. In order to improve the safety profile against these cells we develop and test a novel CAR specific for the CD123 antigens. Here we describe the in vitro and the in vivo efficacy and the safety of this approach based on CIK cells genetically modified to express CAR molecules specific for the CD33 or CD123 antigen. The development and the optimization of the proposed strategy could be a good potential therapeutic tool in the context of minimal residual disease in high-risk transplanted AML patients. Moreover, CAR approach could be potentially used to treat patients resistant to conventional chemotherapeutic approaches or for whom high dose chemotherapy treatment could not be proposed.
- Published
- 2013
24. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor
- Author
-
Ettore Biagi, Chiara F. Magnani, Greta Maria Paola Giordano Attianese, Angel F. Lopez, Virna Marin, Irene Pizzitola, Sarah Tettamanti, Elisabetta Cribioli, Francesca Maltese, Andrea Biondi, Dominique Bonnet, Stefania Galimberti, Tettamanti, S, Marin, V, Pizzitola, I, Magnani, C, GIORDANO ATTIANESE, G, Cribioli, E, Maltese, F, Galimberti, S, Lopez, A, Biondi, A, Bonnet, D, and Biagi, E
- Subjects
AML, CD123, Chimeric Antigen Receptor ,Male ,medicine.medical_treatment ,Recombinant Fusion Proteins ,CD34 ,Interleukin-3 Receptor alpha Subunit ,Receptors, Cell Surface ,Biology ,Immunotherapy, Adoptive ,Leukemia, Myelomonocytic, Acute ,Monocytes ,Cytokine-Induced Killer Cells ,Antigen ,Transduction, Genetic ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Human Umbilical Vein Endothelial Cells ,Humans ,Progenitor cell ,Tumor Stem Cell Assay ,Cytokine-induced killer cell ,Endothelial Cells ,Hematology ,Immunotherapy ,Cytotoxicity Tests, Immunologic ,Hematopoietic Stem Cells ,Chimeric antigen receptor ,Coculture Techniques ,Haematopoiesis ,Leukemia, Myeloid, Acute ,HEK293 Cells ,Immunology ,Leukemia, Monocytic, Acute ,Cytokines ,Female ,Stem cell - Abstract
Summary 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.
- Published
- 2012
25. In Vitro and In Vivo Antitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL against CD33+ Acute Myeloid Leukemia
- Author
-
A. Dutour, V. Marin, I. Pizzitola, S. Valsesia-Wittmann, D. Lee, E. Yvon, H. Finney, A. Lawson, M. Brenner, A. Biondi, E. Biagi, R. Rousseau, Dutour, A, Marin, V, Pizzitola, I, Valsesia Wittmann, S, Lee, D, Yvon, E, Finney, H, Lawson, A, Brenner, M, Biondi, A, Biagi, E, and Rousseau, R
- Subjects
Article Subject ,hemic and lymphatic diseases ,AML, Chimeric Artificial Receptor, CD33, Immunotherapy, Cell Therapy, Gene Therapy ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Genetic engineering of T cells with chimeric T-cell receptors (CARs) is an attractive strategy to treat malignancies. It extends the range of antigens for adoptive T-cell immunotherapy, and major mechanisms of tumor escape are bypassed. With this strategy we redirected immune responses towards the CD33 antigen to target acute myeloid leukemia. To improve in vivo T-cell persistence, we modified human Epstein Barr Virus-(EBV-) specific cytotoxic T cells with an anti-CD33.CAR. Genetically modified T cells displayed EBV and HLA-unrestricted CD33 bispecificity in vitro. In addition, though showing a myeloablative activity, they did not irreversibly impair the clonogenic potential of normal CD34(+) hematopoietic progenitors. Moreover, after intravenous administration into CD33(+) human acute myeloid leukemia-bearing NOD-SCID mice, anti-CD33-EBV-specific T cells reached the tumor sites exerting antitumor activity in vivo. In conclusion, targeting CD33 by CAR-modified EBV-specific T cells may provide additional therapeutic benefit to AML patients as compared to conventional chemotherapy or transplantation regimens alone.
- Published
- 2012
- Full Text
- View/download PDF
26. In vitro comparison of three different chimeric receptor-modified effector T-cell populations for leukemia cell therapy
- Author
-
Raphael Rousseau, Helene Margaret Finney, Alastair D. G. Lawson, Elisabetta Cribioli, Andrea Biondi, Virna Marin, Ettore Biagi, Irene Pizzitola, Martin Pule, Valentina Agostoni, Pizzitola, I, Agostoni, V, Cribioli, E, Pule, M, Rousseau, R, Finney, H, Lawson, A, Biondi, A, Biagi, E, and Marin, V
- Subjects
Cancer Research ,Herpesvirus 4, Human ,Stromal cell ,CD3 Complex ,T cell ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Genetic Vectors ,Cell- and Tissue-Based Therapy ,Receptors, Antigen, T-Cell ,HL-60 Cells ,Biology ,Lymphocyte Activation ,Chimerism ,Viral vector ,Cell therapy ,Antigen ,Transduction, Genetic ,Cell Line, Tumor ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,Child ,Pharmacology ,Leukemia ,Effector ,Immunotherapy ,leukemia immunotherapy, chimeric receptor, cell therapy, gene therapy ,Chemotaxis, Leukocyte ,medicine.anatomical_structure ,Cancer research ,K562 Cells ,T-Lymphocytes, Cytotoxic - 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- Chromium-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 CD33cells 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. © 2011 by Lippincott Williams & Wilkins.
- Published
- 2011
27. Cytokine Induced Killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors
- Author
-
Greta Maria Paola Giordano Attianese, Virna Marin, Valentina Agostoni, Helene Margaret Finney, Raphael Rousseau, Alastair David Griffiths Lawson, Irene Pizzitola, Ettore Biagi, Martin Pule, Andrea Biondi, Marin, V, Pizzitola, I, Agostoni, V, Giordano Attianese, G, Finney, H, Lawson, A, Pule, M, Rousseau, R, Biondi, A, and Biagi, E
- Subjects
Cytotoxicity, Immunologic ,Time Factors ,Recombinant Fusion Proteins ,medicine.medical_treatment ,CIK, CD33, Chimeric Receptor, gene therapy ,Sialic Acid Binding Ig-like Lectin 3 ,CD33 ,Cell- and Tissue-Based Therapy ,Receptors, Cytoplasmic and Nuclear ,HL-60 Cells ,Biology ,Cytokine-Induced Killer Cells ,Cell Line, Tumor ,medicine ,Humans ,CD135 ,Child ,Cells, Cultured ,Cell Proliferation ,Lymphokine-activated killer cell ,Cytokine-induced killer cell ,Myeloid leukemia ,Hematology ,Flow Cytometry ,Natural killer T cell ,Coculture Techniques ,Haematopoiesis ,HEK293 Cells ,Cytokine ,Leukemia, Myeloid ,Acute Disease ,Immunology ,Original Article - Abstract
Background. Cytokine induced killer cells are ex-vivo expanded cells with potent antitumoral activity. Cytokine induced killer cells infusion in acute myeloid leukemia patients relapsing after allogeneic hematopoietic stem cell transplant is well tolerated, but limited clinical responses were 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 in vitro characterized for their ability to lyse leukemic targets (4-hour-51Chromium-release and 6-day co-cultures assays on human stromal mesenchymal cells), to proliferate (3H-thymidine-incorporation assay) and to secrete cytokines (Flow cytomix assay) after contact with acute myeloid leukemia cells. Their activity against normal CD34+ hematopoietic progenitors 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 a prominent CD33-specific proliferative activity, with a release of high levels of immunostimulatory cytokines. The presence of CD28-OX40 in chimeric receptor endodomain was associated with a significant amelioration of cytokine induced killer cells anti-leukemic activity. Importantly, even though anti-CD33 chimeric receptors-transduced cytokine induced killer cells showed toxicity against normal hematopoietic CD34+ progenitors, a residual clonogenic activity was preserved. Conclusions. Our results indicate that anti-CD33 chimeric receptors strongly enhance anti-leukemic cytokine induced killer functions, suggesting that cytokine induced killer cells transduced with these molecules might represent a promising optimized tool for acute myeloid leukemia immunotherapy.
- Published
- 2010
28. Chimeric antigen receptor for specific targeting of acute myeloid leukemia
- Author
-
Sarah Tettamanti, Dominique Bonnet, Irene Pizzitola, Andrea Biondi, Francois Lassailly, Kevin Rouault-Pierre, Fernando Anjos-Afonso, Ettore Biagi, Pizzitola, I, Anjos Afonso, F, Rouault Pierre, K, Lassailly, F, Tettamanti, S, Biondi, A, Biagi, E, and Bonnet, D
- Subjects
Acute Myeloid Leukemia ,Cancer Research ,T cell ,CD33 ,Immunology ,CD34 ,Biology ,Biochemistry ,Antigen ,medicine ,Immunology and Allergy ,CD135 ,Genetics (clinical) ,Transplantation ,RUNX1T1 ,Myeloid leukemia ,Hematology ,Cell Biology ,Minimal residual disease ,Chimeric antigen receptor ,Haematopoiesis ,medicine.anatomical_structure ,Oncology ,Cord blood ,Cancer research ,Bone marrow ,Chimeric Antigen Receptor - Abstract
Abstract 4225 Despite the progress in the treatment of acute myeloid leukemia (AML) achieved in the last decades, a significant number of patients are still refractory to or relapse after conventional chemotherapy regimens. Therefore it is necessary to develop novel alternative approaches. Immunotherapy with T cells genetically modified to express chimeric antigen receptors (CARs) represent a valid option in this sense. CARs are artificial T-cell receptors constituted by a specific antigen-binding domain, and a signaling region, that, upon antigen recognition, leads to T-cell activation, and lysis of the target cells. AML is a potential optimal target for CAR strategy because of the over-expression of a number of surface antigens like CD33, CD123. Since CD33 is also expressed on normal hematopoietic stem/progenitors cells (HSPCs) resulting in a potential severe impairment of normal myelopoiesis, CD123 has recently emerged as new potential attractive molecules based on its differential expression pattern, being still wildly overexpressed by AML population, and at the same time less expressed on HSPCs. Here we describe the in vivo efficacy and the safety of this approach based on Cytokine-Induced-Killers (CIK) cells genetically modified to express CAR molecules specific for the CD33 or CD123 antigen. Once injected into low-level AML engrafted NSG mice (median of hCD45+CD33+ 0.6% before treatment), genetically modify T cells had a potent antitumor effect. Indeed, the bone marrow of control untreated animals or mice treated with un-manipulated CIK cells, was infiltrated by leukemic cells (86% and 81% leukemic engraftment), while in 7/8 anti-CD33-CD28-OX40-ζ and 8/10 anti-CD123-CD28-OX40-ζ treated mice we couldn't detect any AML cells. Similar results have been obtained when the treatment via T cell injection start when high AML burden has been obtained (median of hCD45+CD33+ 70% before treatment). One week after the last CIK's injection the level of AML engraftment was 96%, 87%, 0.35% and 0.34% for untreated mice, mice treated with un-manipulated CIK cells and with anti-CD33-CD28-OX40-ζ and anti-CD123-CD28-OX40-ζ transduced CIK-cells respectively. We performed secondary transplantation on the residual AML cells present in these animals and mice were treated again with transduced CIK cells. Residual AML cells were still sensitive to CARs approach, leading once again to an almost complete eradication of the disease (median level of hCD45+CD33+ engraftment was 98%, 0.02% and 0.04% respectively for untreated mice, anti-CD33-CD28-OX40-ζ and anti-CD123-CD28-OX40-ζ transduced CIK-cells). Furthermore, a fundamental issue was to determine the safety profile of such approach against normal hematopoietic precursors. In untreated mice injected with primary cord blood derived CD34+ cells the level of engraftment of hCD45 compartment was 42% whilst in mice treated with un-manipulated, anti-CD33-CD28-OX40-ζ or anti-CD123-CD28-OX40-ζ transduced CIK-cells the levels of human compartment was 40%, 11.7% and 26.3% respectively. Moreover when we consider specifically the CD34+CD38- compartment, enriched in HSC, the level of engraftment was 1.92%, 1.02%, 0.55% and 0.83%. Secondary transplantations are now ongoing to give a more complete profile about the remaining HSC repopulating capability after treatment. To more closely mimic a physiological context, similar experiments are ongoing using mice engrafted with normal adult bone marrow instead of umbilical cord blood. These experiments should offer relevant information concerning the efficacy and safety of the proposed strategy particularly in the context of minimal residual disease in high-risk transplanted AML patients. Moreover CAR approach could be potentially used to treat patients resistant to conventional chemotherapeutic approaches or for whom high dose chemotherapy treatment could not be proposed. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
- Full Text
- View/download PDF
29. New advances in leukaemia immunotherapy by the use of Chimeric Artificial Antigen Receptors (CARs): state of the art and perspectives for the near future
- Author
-
Irene Pizzitola, Andrea Biondi, Greta Maria Paola Giordano Attianese, Ettore Biagi, Virna Marin, Sarah Tettamanti, Elisabetta Cribioli, Biagi, E, Marin, V, Giordano Attianese, G, Pizzitola, I, Tettamanti, S, Cribioli, E, and Biondi, A
- Subjects
Recombinant Fusion Proteins ,Genetic enhancement ,medicine.medical_treatment ,Receptors, Antigen, T-Cell ,Review ,Immunotherapy, Adoptive ,Cell therapy ,Immune system ,medicine ,Humans ,Leukemia ,business.industry ,T-cell receptor ,lcsh:RJ1-570 ,lcsh:Pediatrics ,Immunotherapy ,medicine.disease ,gene therapy ,Peptide Fragments ,Chimeric antigen receptor ,Leukemia, Lymphoid ,chimeric antigen receptor, adoptive immunotherapy, bone marrow transplantation, leukemia ,Leukemia, Myeloid ,chimeric artificial receptors ,Immunology ,Cancer research ,cell therapy ,Signal transduction ,business ,Signal Transduction ,Leukaemia immunotherapy - 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.
- Full Text
- View/download PDF
30. γ-Catenin-Dependent Signals Maintain BCR-ABL1 + B Cell Acute Lymphoblastic Leukemia.
- Author
-
Luong-Gardiol N, Siddiqui I, Pizzitola I, Jeevan-Raj B, Charmoy M, Huang Y, Irmisch A, Curtet S, Angelov GS, Danilo M, Juilland M, Bornhauser B, Thome M, Hantschel O, Chalandon Y, Cazzaniga G, Bourquin JP, Huelsken J, and Held W
- Subjects
- 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, Fusion Proteins, bcr-abl metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Wnt Signaling Pathway, gamma Catenin metabolism
- 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., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
31. Tumour-derived PGD2 and NKp30-B7H6 engagement drives an immunosuppressive ILC2-MDSC axis.
- Author
-
Trabanelli S, Chevalier MF, Martinez-Usatorre A, Gomez-Cadena A, Salomé B, Lecciso M, Salvestrini V, Verdeil G, Racle J, Papayannidis C, Morita H, Pizzitola I, Grandclément C, Bohner P, Bruni E, Girotra M, Pallavi R, Falvo P, Leibundgut EO, Baerlocher GM, Carlo-Stella C, Taurino D, Santoro A, Spinelli O, Rambaldi A, Giarin E, Basso G, Tresoldi C, Ciceri F, Gfeller D, Akdis CA, Mazzarella L, Minucci S, Pelicci PG, Marcenaro E, McKenzie ANJ, Vanhecke D, Coukos G, Mavilio D, Curti A, Derré L, and Jandus C
- Subjects
- 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, B7 Antigens immunology, Lymphocytes immunology, Myeloid-Derived Suppressor Cells immunology, Natural Cytotoxicity Triggering Receptor 3 immunology, Prostaglandin D2 immunology
- 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.
- Published
- 2017
- Full Text
- View/download PDF
32. Mechanisms of efficacy in cancer immunotherapy: 14th Annual Meeting of the Association for Cancer Immunotherapy (CIMT), Mainz, Germany, May 10-12, 2016.
- Author
-
Theelen J, Braun J, Hörzer H, Kloke BP, Ott M, Pizzitola I, Schirmer U, and Miller M
- Subjects
- Cancer Vaccines, Cell- and Tissue-Based Therapy, Humans, Immunomodulation, Molecular Targeted Therapy, Neoplasms pathology, Tumor Microenvironment immunology, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy
- Published
- 2017
- Full Text
- View/download PDF
33. Myelodysplastic syndrome can propagate from the multipotent progenitor compartment.
- Author
-
Rouault-Pierre K, Smith AE, Mian SA, Pizzitola I, Kulasekararaj AG, Mufti GJ, and Bonnet D
- Subjects
- Humans, Male, Middle Aged, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, Myelodysplastic Syndromes metabolism, Myelodysplastic Syndromes pathology, Stem Cell Niche
- Published
- 2017
- Full Text
- View/download PDF
34. Phage Selection of Chemically Stabilized α-Helical Peptide Ligands.
- Author
-
Diderich P, Bertoldo D, Dessen P, Khan MM, Pizzitola I, Held W, Huelsken J, and Heinis C
- Subjects
- Amino Acid Sequence, Molecular Conformation, Molecular Sequence Data, Peptide Library, Peptides chemistry, beta Catenin metabolism, Bacteriophages metabolism, Ligands
- 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 β-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.
- Published
- 2016
- Full Text
- View/download PDF
35. SF3B1 mutant MDS-initiating cells may arise from the haematopoietic stem cell compartment.
- Author
-
Mian SA, Rouault-Pierre K, Smith AE, Seidl T, Pizzitola I, Kizilors A, Kulasekararaj AG, Bonnet D, and Mufti GJ
- Subjects
- 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, 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
- 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.
- Published
- 2015
- Full Text
- View/download PDF
36. A niche-like culture system allowing the maintenance of primary human acute myeloid leukemia-initiating cells: a new tool to decipher their chemoresistance and self-renewal mechanisms.
- Author
-
Griessinger E, Anjos-Afonso F, Pizzitola I, Rouault-Pierre K, Vargaftig J, Taussig D, Gribben J, Lassailly F, and Bonnet D
- Subjects
- 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, Antimetabolites, Antineoplastic pharmacology, Cytarabine pharmacology, Drug Resistance, Neoplasm drug effects, Leukemia, Myeloid, Acute drug therapy, Neoplastic Stem Cells metabolism, Stem Cell Niche
- 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.
- Published
- 2014
- Full Text
- View/download PDF
37. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor.
- Author
-
Tettamanti S, Marin V, Pizzitola I, Magnani CF, Giordano Attianese GM, Cribioli E, Maltese F, Galimberti S, Lopez AF, Biondi A, Bonnet D, and Biagi E
- Subjects
- 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, Cytokine-Induced Killer Cells immunology, Immunotherapy, Adoptive methods, Leukemia, Myeloid, Acute pathology, Leukemia, Myelomonocytic, Acute pathology, Receptors, Cell Surface physiology
- 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., (© 2013 Blackwell Publishing Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
38. Upregulation of adhesion molecules on leukemia targets improves the efficacy of cytotoxic T cells transduced with chimeric anti-CD19 receptor.
- Author
-
Laurin D, Marin V, Biagi E, Pizzitola I, Agostoni V, Gallot G, Vié H, Jacob MC, Chaperot L, Aspord C, and Plumas J
- Subjects
- 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, Antigens, CD19 immunology, Intercellular Adhesion Molecule-1 metabolism, Leukemia, B-Cell immunology, Receptors, Antigen, B-Cell immunology, T-Lymphocytes, Cytotoxic immunology
- 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.
- Published
- 2013
- Full Text
- View/download PDF
39. Comparison of different suicide-gene strategies for the safety improvement of genetically manipulated T cells.
- Author
-
Marin V, Cribioli E, Philip B, Tettamanti S, Pizzitola I, Biondi A, Biagi E, and Pule M
- Subjects
- 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, Caspase 9 genetics, Genes, Transgenic, Suicide genetics, T-Lymphocytes, Cytotoxic metabolism, Thymidine Kinase genetics
- 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.
- Published
- 2012
- Full Text
- View/download PDF
40. In Vitro and In Vivo Antitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL against CD33 Acute Myeloid Leukemia.
- Author
-
Dutour A, Marin V, Pizzitola I, Valsesia-Wittmann S, Lee D, Yvon E, Finney H, Lawson A, Brenner M, Biondi A, Biagi E, and Rousseau R
- Abstract
Genetic engineering of T cells with chimeric T-cell receptors (CARs) is an attractive strategy to treat malignancies. It extends the range of antigens for adoptive T-cell immunotherapy, and major mechanisms of tumor escape are bypassed. With this strategy we redirected immune responses towards the CD33 antigen to target acute myeloid leukemia. To improve in vivo T-cell persistence, we modified human Epstein Barr Virus-(EBV-) specific cytotoxic T cells with an anti-CD33.CAR. Genetically modified T cells displayed EBV and HLA-unrestricted CD33 bispecificity in vitro. In addition, though showing a myeloablative activity, they did not irreversibly impair the clonogenic potential of normal CD34(+) hematopoietic progenitors. Moreover, after intravenous administration into CD33(+) human acute myeloid leukemia-bearing NOD-SCID mice, anti-CD33-EBV-specific T cells reached the tumor sites exerting antitumor activity in vivo. In conclusion, targeting CD33 by CAR-modified EBV-specific T cells may provide additional therapeutic benefit to AML patients as compared to conventional chemotherapy or transplantation regimens alone.
- Published
- 2012
- Full Text
- View/download PDF
41. New advances in leukaemia immunotherapy by the use of Chimeric Artificial Antigen Receptors (CARs): state of the art and perspectives for the near future.
- Author
-
Biagi E, Marin V, Attianese GM, Pizzitola I, Tettamanti S, Cribioli E, and Biondi A
- Subjects
- Humans, Immunotherapy, Adoptive methods, Leukemia, Lymphoid therapy, Leukemia, Myeloid therapy, Peptide Fragments immunology, Receptors, Antigen, T-Cell genetics, Recombinant Fusion Proteins genetics, Signal Transduction, Immunotherapy, Adoptive trends, Leukemia therapy, Receptors, Antigen, T-Cell immunology, Recombinant Fusion Proteins immunology
- 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.
- Published
- 2011
- Full Text
- View/download PDF
42. In vitro comparison of three different chimeric receptor-modified effector T-cell populations for leukemia cell therapy.
- Author
-
Pizzitola I, Agostoni V, Cribioli E, Pule M, Rousseau R, Finney H, Lawson A, Biondi A, Biagi E, and Marin V
- Subjects
- 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, T-Lymphocytes, Cytotoxic immunology, Transduction, Genetic, Cell- and Tissue-Based Therapy, Chimerism, Leukemia immunology, Leukemia therapy, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes immunology, T-Lymphocytes metabolism
- 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.
- Published
- 2011
- Full Text
- View/download PDF
43. In vitro and in vivo model of a novel immunotherapy approach for chronic lymphocytic leukemia by anti-CD23 chimeric antigen receptor.
- Author
-
Giordano Attianese GM, Marin V, Hoyos V, Savoldo B, Pizzitola I, Tettamanti S, Agostoni V, Parma M, Ponzoni M, Bertilaccio MT, Ghia P, Biondi A, Dotti G, and Biagi E
- Subjects
- 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, Immunotherapy, Adoptive methods, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Receptors, IgE antagonists & inhibitors
- 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-β, 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.
- Published
- 2011
- Full Text
- View/download PDF
44. Exploration of the lysis mechanisms of leukaemic blasts by chimaeric T-cells.
- Author
-
Laurin D, Marin V, Biagi E, Pizzitola I, Agostoni V, Gallot G, Vié H, Jacob MC, Chaperot L, Aspord C, and Plumas J
- Subjects
- 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, Apoptosis, Cytokine-Induced Killer Cells metabolism, Immunotherapy, Adoptive methods, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, T-Lymphocytes, Cytotoxic metabolism
- 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.
- Published
- 2010
- Full Text
- View/download PDF
45. Deficiency of the long pentraxin PTX3 promotes vascular inflammation and atherosclerosis.
- Author
-
Norata GD, Marchesi P, Pulakazhi Venu VK, Pasqualini F, Anselmo A, Moalli F, Pizzitola I, Garlanda C, Mantovani A, and Catapano AL
- Subjects
- 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, Atherosclerosis genetics, Atherosclerosis immunology, C-Reactive Protein genetics, C-Reactive Protein immunology, Nerve Tissue Proteins genetics, Nerve Tissue Proteins immunology, Vasculitis genetics, Vasculitis immunology
- 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.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.