46 results on '"Chiara Buracchi"'
Search Results
2. Phenotypic profiling of CD34+ cells by advanced flow cytometry improves diagnosis of juvenile myelomonocytic leukemia
- Author
-
Cristina Bugarin, Laura Antolini, Chiara Buracchi, Sergio Matarraz, Tiziana Angela Coliva, Vincent H. van der Velden, Tomasz Szczepanski, Elaine Sobral da Costa, Alita van der Sluijs, Michaela Novakova, Ester Mejstrikova, Stefan Nierkens, Fabiana Vieira de Mello, Paula Fernandez, Carmen Aanei, Łukasz Sędek, Luisa Strocchio, Riccardo Masetti, Laura Sainati, Jan Philippé, Maria Grazia Valsecchi, Franco Locatelli, Jacques J.M. van Dongen, Andrea Biondi, Alberto Orfao, and Giuseppe Gaipa
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Diagnostic criteria for juvenile myelomonocytic leukemia (JMML) are currently well defined, however in some patients diagnosis still remains a challenge. Flow cytometry is a well established tool for diagnosis and follow-up of hematological malignancies, nevertheless it is not routinely used for JMML diagnosis. Herewith, we characterized the CD34+ hematopoietic precursor cells collected from 31 children with JMML using a combination of standardized EuroFlow antibody panels to assess the ability to discriminate JMML cells from normal/reactive bone marrow cell as controls (n=29) or from cells of children with other hematological diseases mimicking JMML (n=9). CD34+ precursors in JMML showed markedly reduced B-cell and erythroid-committed precursors compared to controls, whereas monocytic and CD7+ lymphoid precursors were significantly expanded. Moreover, aberrant immunophenotypes were consistently present in CD34+ precursors in JMML, while they were virtually absent in controls. Multivariate logistic regression analysis showed that combined assessment of the number of CD34+CD7+ lymphoid precursors and CD34+ aberrant precursors or erythroid precursors had a great potential in discriminating JMMLs versus controls. Importantly our scoring model allowed highly efficient discrimination of truly JMML versus patients with JMML-like diseases. In conclusion, we show for the first time that CD34+ precursors from JMML patients display a unique immunophenotypic profile which might contribute to a fast and accurate diagnosis of JMML worldwide by applying an easy to standardize single eight-color antibody combination.
- Published
- 2023
- Full Text
- View/download PDF
3. Therapeutic afucosylated monoclonal antibody and bispecific T-cell engagers for T-cell acute lymphoblastic leukemia
- Author
-
Daniele Caracciolo, Antonio Giordano, Pierfrancesco Tassone, Caterina Riillo, Andrea Ballerini, Giuseppe Gaipa, Ludovic Lhermitte, Marco Rossi, Eugénie Duroyon, Katia Grillone, Maria Eugenia Gallo Cantafio, Chiara Buracchi, Greta Alampi, Alessandro Gulino, Beatrice Belmonte, Francesco Conforti, Gaetanina Golino, Giada Juli, Emanuela Altomare, Nicoletta Polerà, Francesca Scionti, Mariamena Arbitrio, Michelangelo Iannone, Massimo Martino, Gabriella Talarico, Andrea Ghelli Luserna di Rorà, Anna Ferrari, Simona Sestito, Licia Pensabene, Markus Hildinger, Maria Teresa Di Martino, Giovanni Martinelli, Claudio Tripodo, and Vahid Asnafi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease with a poor cure rate for relapsed/resistant patients. Due to the lack of T-cell restricted targetable antigens, effective immune-therapeutics are not presently available and the treatment of chemo-refractory T-ALL is still an unmet clinical need. To develop novel immune-therapy for T-ALL, we generated an afucosylated monoclonal antibody (mAb) (ahuUMG1) and two different bispecific T-cell engagers (BTCEs) against UMG1, a unique CD43-epitope highly and selectively expressed by T-ALL cells from pediatric and adult patients.Methods UMG1 expression was assessed by immunohistochemistry (IHC) on a wide panel of normal tissue microarrays (TMAs), and by flow cytometry on healthy peripheral blood/bone marrow-derived cells, on 10 different T-ALL cell lines, and on 110 T-ALL primary patient-derived cells. CD43-UMG1 binding site was defined through a peptide microarray scanning. ahuUMG1 was generated by Genetic Glyco-Engineering technology from a novel humanized mAb directed against UMG1 (huUMG1). BTCEs were generated as IgG1-(scFv)2 constructs with bivalent (2+2) or monovalent (2+1) CD3ε arms. Antibody dependent cellular cytotoxicity (ADCC), antibody dependent cellular phagocytosis (ADCP) and redirected T-cell cytotoxicity assays were analysed by flow cytometry. In vivo antitumor activity of ahUMG1 and UMG1-BTCEs was investigated in NSG mice against subcutaneous and orthotopic xenografts of human T-ALL.Results Among 110 T-ALL patient-derived samples, 53 (48.1%) stained positive (24% of TI/TII, 82% of TIII and 42.8% of TIV). Importantly, no expression of UMG1-epitope was found in normal tissues/cells, excluding cortical thymocytes and a minority (2 log lower as compared with ahuUMG1, with significant mice survival advantage in different T-ALL models in vivo.Conclusion Altogether our findings, including the safe UMG1-epitope expression profile, provide a framework for the clinical development of these innovative immune-therapeutics for this still orphan disease.
- Published
- 2021
- Full Text
- View/download PDF
4. Targeted Gene Correction in Osteopetrotic-Induced Pluripotent Stem Cells for the Generation of Functional Osteoclasts
- Author
-
Tui Neri, Sharon Muggeo, Marianna Paulis, Maria Elena Caldana, Laura Crisafulli, Dario Strina, Maria Luisa Focarelli, Francesca Faggioli, Camilla Recordati, Samantha Scaramuzza, Eugenio Scanziani, Stefano Mantero, Chiara Buracchi, Cristina Sobacchi, Angelo Lombardo, Luigi Naldini, Paolo Vezzoni, Anna Villa, and Francesca Ficara
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Autosomal recessive osteopetrosis is a human bone disease mainly caused by TCIRG1 gene mutations that prevent osteoclasts resorbing activity, recapitulated by the oc/oc mouse model. Bone marrow transplantation is the only available treatment, limited by the need for a matched donor. The use of induced pluripotent stem cells (iPSCs) as an unlimited source of autologous cells to generate gene corrected osteoclasts might represent a powerful alternative. We generated iPSCs from oc/oc mice, corrected the mutation using a BAC carrying the entire Tcirg1 gene locus as a template for homologous recombination, and induced hematopoietic differentiation. Similarly to physiologic fetal hematopoiesis, iPSC-derived CD41+ cells gradually gave rise to CD45+ cells, which comprised both mature myeloid cells and high proliferative potential colony-forming cells. Finally, we differentiated the gene corrected iPSC-derived myeloid cells into osteoclasts with rescued bone resorbing activity. These results are promising for a future translation into the human clinical setting.
- Published
- 2015
- Full Text
- View/download PDF
5. Long-Term Host Immune Modulation Following Tisagenlecleucel Administration in Patients with Diffuse Large B-Cell Lymphoma and B-Lineage Acute Lymphoblastic Leukemia
- Author
-
Anna Guarini, Giulia Radice, Nadia Peragine, Chiara Buracchi, Maria Stefania De Propris, Alice Di Rocco, Arianna Di Rocco, Sabina Chiaretti, Alex Moretti, Sara Napolitano, Maurizio Martelli, Adriana Balduzzi, Giuseppe Gaipa, Andrea Biondi, Robin Foà, Guarini, A, Radice, G, Peragine, N, Buracchi, C, De Propris, M, Di Rocco, A, Chiaretti, S, Moretti, A, Napolitano, S, Martelli, M, Balduzzi, A, Gaipa, G, Biondi, A, and Foà, R
- Subjects
Cancer Research ,CAR-T cells ,DLBCL ,B-ALL ,immune modulation ,Oncology ,CAR-T cell - Abstract
Background: Chimeric antigen receptor (CAR)-T cells represent a potentially curative strategy for patients with relapsed or refractory (R/R) B-cell malignancies. To elucidate a possible host immune activation following CAR-T-cell infusion, we investigated the effects of tisagenlecleucel administration on the patients’ immune populations in 25 patients with R/R diffuse large B-cell lymphoma (DLBCL) and B-lineage acute lymphoblastic leukemia (B-ALL). Methods: The modulation of CAR-T cells over time, the numeric changes, as well as the cytokine production capability of different lymphocyte populations and circulating cytokine levels, were analyzed. Results: Our results confirmed the ability of tisagenlecleucel to control the disease, with an overall response observed in 84.6% of DLBCL and in 91.7% of B-ALL patients at 1-month post-infusion, and showed that most patients who subsequently relapsed could undergo further treatment. Interestingly, we could document a significant increase in CD3+, CD4+, CD8+, and NK cells over time, as well as a decrease in Treg cells, and an increased IFNγ and TNFα production by T lymphocytes. Conclusions: Taken together, our results indicate that in patients with DLBCL and B-ALL, the administration of tisagenlecleucel is capable of inducing a marked and prolonged in vivo modulation/reshaping of the host immune system, both in children and adults.
- Published
- 2023
6. First Description of a Frameshift PAX5 Germline Variant in Two Siblings with B-Cell Precursor Acute Lymphoblastic Leukemia
- Author
-
Laura Rachele Bettini, Grazia Fazio, Claudia Saitta, Sonia Palamini, Chiara Buracchi, Stefano Rebellato, Nicola Santoro, Cristiano Simone, Andrea Biondi, and Giovanni Cazzaniga
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
7. Final Results of Phase I/II Study of Donor-Derived CAR T Cells Engineered with Sleeping Beauty in Pediatric and Adult Patients with B-Cell Acute Lymphoblastic Leukemia Relapsed Post-HSCT
- Author
-
Federico Lussana, Chiara Francesca Magnani, Giuseppe Gaipa, Stefania Galimberti, Giuseppe Gritti, Daniela Belotti, Sara Napolitano, Chiara Buracchi, Gian Maria Borleri, Benedetta Rambaldi, Giuliana Rizzuto, Anna Grassi, Muriel Paganessi, Silvia Ferrari, Sarah Tettamanti, Giovanni Gazzaniga, Chiara Capelli, Elisa Gotti, Martino Introna, Adriana Balduzzi, Maria Grazia Valsecchi, Giuseppe Dastoli, Alessandro Rambaldi, and Andrea Biondi
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
8. Carcik-CD19 Cells Expand In Vivo Toward a CD8+ Memory Phenotype and Their Persistence Is Associated with a Longer Duration of Response
- Author
-
Benedetta Rambaldi, Stefania Galimberti, Giuliana Rizzuto, Chiara Francesca Magnani, Chiara Buracchi, Giulia Risca, Martina Paredi, Daniela Belotti, Alex Moretti, Marianna Ponzo, Sarah Tettamanti, Gian Maria Borleri, Cristian Meli, Muriel Paganessi, Silvia Zaninelli, Elisa Gotti, Chiara Capelli, Martino Introna, Federico Lussana, Giuseppe Gritti, Silvia Ferrari, Anna Grassi, Sara Napolitano, Adriana Balduzzi, Maria Grazia Valsecchi, Giuseppe Dastoli, Alessandro Rambaldi, Andrea Biondi, and Giuseppe Gaipa
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
9. Sleeping Beauty–engineered CAR T cells achieve antileukemic activity without severe toxicities
- Author
-
Gianmaria Borleri, Adriana Balduzzi, Benedetta Cabiati, Michele Quaroni, Martino Introna, Grazia Fazio, Sara Napolitano, Chiara Buracchi, Stefania Cesana, Giovanni Cazzaniga, Giuseppe Gaipa, Giuseppe Gritti, Giada Matera, Silvia Zaninelli, Ettore Biagi, Andrea Biondi, Stefania Galimberti, Federico Lussana, Fabrizio Benedicenti, Attilio Rovelli, Giuseppe Dastoli, Eugenio Montini, Sarah Tettamanti, Valentina Colombo, Andrea Calabria, Maria Grazia Valsecchi, Alessandro Rambaldi, Silvia Ferrari, Chiara F. Magnani, Daniela Belotti, Magnani, C, Gaipa, G, Lussana, F, Belotti, D, Gritti, G, Napolitano, S, Matera, G, Cabiati, B, Buracchi, C, Borleri, G, Fazio, G, Zaninelli, S, Tettamanti, S, Cesana, S, Colombo, V, Quaroni, M, Cazzaniga, G, Rovelli, A, Biagi, E, Galimberti, S, Calabria, A, Benedicenti, F, Montini, E, Ferrari, S, Introna, M, Balduzzi, A, Valsecchi, M, Dastoli, G, Rambaldi, A, and Biondi, A
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Adolescent ,CD3 ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Immunotherapy, Adoptive ,CD19 ,03 medical and health sciences ,0302 clinical medicine ,Refractory ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Leukemias ,medicine ,Humans ,Clinical Trials ,Child ,Hematology ,biology ,Cancer gene therapy ,business.industry ,Hematopoietic Stem Cell Transplantation ,Infant ,Cancer ,General Medicine ,Immunotherapy ,Allografts ,medicine.disease ,Clinical trial ,030104 developmental biology ,Child, Preschool ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Clinical Medicine ,business - Abstract
BACKGROUND: Chimeric antigen receptor (CAR) T cell immunotherapy has resulted in complete remission (CR) and durable response in highly refractory patients. However, logistical complexity and high costs of manufacturing autologous viral products limit CAR T cell availability. METHODS: We report the early results of a phase I/II trial in B cell acute lymphoblastic leukemia (B-ALL) patients relapsed after allogeneic hematopoietic stem cell transplantation (HSCT) using donor-derived CD19 CAR T cells generated with the Sleeping Beauty (SB) transposon and differentiated into cytokine-induced killer (CIK) cells. RESULTS: The cellular product was produced successfully for all patients from the donor peripheral blood (PB) and consisted mostly of CD3(+) lymphocytes with 43% CAR expression. Four pediatric and 9 adult patients were infused with a single dose of CAR T cells. Toxicities reported were 2 grade I and 1 grade II cytokine-release syndrome (CRS) cases at the highest dose in the absence of graft-versus-host disease (GVHD), neurotoxicity, or dose-limiting toxicities. Six out of 7 patients receiving the highest doses achieved CR and CR with incomplete blood count recovery (CRi) at day 28. Five out of 6 patients in CR were also minimal residual disease negative (MRD(–)). Robust expansion was achieved in the majority of the patients. CAR T cells were measurable by transgene copy PCR up to 10 months. Integration site analysis showed a positive safety profile and highly polyclonal repertoire in vitro and at early time points after infusion. CONCLUSION: SB-engineered CAR T cells expand and persist in pediatric and adult B-ALL patients relapsed after HSCT. Antileukemic activity was achieved without severe toxicities. TRIAL REGISTRATION: ClinicalTrials.gov NCT03389035. FUNDING: This study was supported by grants from the Fondazione AIRC per la Ricerca sul Cancro (AIRC); Cancer Research UK (CRUK); the Fundación Científica de la Asociación Española Contra el Cáncer (FC AECC); Ministero Della Salute; Fondazione Regionale per la Ricerca Biomedica (FRRB).
- Published
- 2020
- Full Text
- View/download PDF
10. Single‐cell profiling of pediatric T‐cell acute lymphoblastic leukemia: Impact of <scp> PTEN </scp> exon 7 mutation on <scp>PI3K</scp> / <scp>Akt</scp> and <scp>JAK–STAT</scp> signaling pathways
- Author
-
Grazia Fazio, Giuseppe Gaipa, Andrea Biondi, Chiara Buracchi, Luca Lo Nigro, Paola Bonaccorso, and Cristina Bugarin
- Subjects
0301 basic medicine ,Cell signaling ,Histology ,biology ,T cell ,Cell Biology ,Pathology and Forensic Medicine ,03 medical and health sciences ,Exon ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,medicine ,Cancer research ,PTEN ,Tensin ,Signal transduction ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
Background The PI3K/Akt/mTOR (PI3K) signaling pathway has a crucial role in T-cell acute lymphoblastic leukemias (T-ALLs). Although loss-of-function of phosphatase and tensin homolog (PTEN) is a common event in pediatric T-ALLs, the exact role of this tumor suppressor in T-ALL development has yet to be defined. Methods Here, we report an optimized cytometric method for accurate proteomic profiling of T-ALL leukemic blasts at single-cell level. We determined the expression of PI3K and JAK-STAT signaling components in both primary and immortalized T-ALL cells as well as in normal T cells. Results We observed that PTEN exon 7 mutated T-ALL cells retain a distinct PI3K activation; in particular, these cells show higher pAkt levels and a lower pS6 expression. Interestingly, we demonstrated for the first time that PTEN exon 7 mutated T-ALL are nonresponsive to IL7 in vitro as assessed by lack of pSTAT5 activation, although they do express IL7R. Conclusions Phosphoflow analysis represents a fast, reliable, and accurate method to study the signaling profile of T-ALL. PTEN exon 7 mutated T-ALL cells are nonresponsive to IL7 in vitro suggesting that they may activate other mechanisms to support their viability and proliferation such as a higher constitutive PI3K/Akt signaling. Further investigations are necessary to elucidate the significance of this peculiar signaling behavior. Our observations should be taken into account in future studies aiming at molecular targeting of PI3K and/or JAK/STAT pathways for pharmacological intervention in T-ALL.
- Published
- 2020
- Full Text
- View/download PDF
11. Impact of Pre-Analytical and Analytical Variables Associated with Sample Preparation on Flow Cytometric Stainings Obtained with EuroFlow Panels
- Author
-
Łukasz Sędek, Juan Flores-Montero, Alita van der Sluijs, Jan Kulis, Jeroen te Marvelde, Jan Philippé, Sebastian Böttcher, Marieke Bitter, Joana Caetano, Vincent H. J. van der Velden, Edwin Sonneveld, Chiara Buracchi, Ana Helena Santos, Margarida Lima, Tomasz Szczepański, Jacques J. M. van Dongen, Alberto Orfao, European Commission, European Hematology Association, Polish National Agency for Academic Exchange, Silesian University of Technology, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Immunology, Sedek, L, Flores-Montero, J, van der Sluijs, A, Kulis, J, Marvelde, J, Philippe, J, Bottcher, S, Bitter, M, Caetano, J, van der Velden, V, Sonneveld, E, Buracchi, C, Santos, A, Lima, M, Szczepanski, T, van Dongen, J, and Orfao, A
- Subjects
standardization ,Cancer Research ,Leukemia ,Lymphoma ,flow cytometry ,anticoagulant ,Anticoagulant ,leukemia ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lymphoma ,Sample storage ,Standardization ,Immunophenotyping ,multiple myeloma ,sample storage ,Oncology ,immunophenotyping ,SDG 3 - Good Health and Well-being ,Multiple myeloma ,Protocol ,Flow cytometry ,protocol ,RC254-282 - Abstract
Objective interpretation of FC results may still be hampered by limited technical standardization. The EuroFlow consortium conducted a series of experiments to determine the impact of different variables on the relative distribution and the median fluorescence intensity (MFI) of markers stained on different cell populations, from both healthy donors and patients’ samples with distinct hematological malignancies. The use of different anticoagulants; the time interval between sample collection, preparation, and acquisition; pH of washing buffers; and the use of cell surface membrane-only (SM) vs. cell surface plus intracytoplasmic (SM+CY) staining protocols, were evaluated. Our results showed that only monocytes were represented at higher percentages in EDTA- vs. heparin-anticoagulated samples. Application of SM or SM+CY protocols resulted in slight differences in the percentage of neutrophils and debris determined only with particular antibody combinations. In turn, storage of samples for 24 h at RT was associated with greater percentage of debris and cell doublets when the plasma cell disorder panel was used. Furthermore, 24 h storage of stained cells at RT was selectively detrimental for MFI levels of CD19 and CD45 on mature B- and T-cells (but not on leukemic blasts, clonal B- and plasma cells, neutrophils, and NK cells). The obtained results showed that the variables evaluated might need to be tailored for sample and cell type(s) as well as to the specific markers compared; however, defining of well-balanced boundaries for storage time, staining-to-acquisition delay, and pH of washing buffer would be a valid recommendation for most applications and circumstances described herein., This research was funded by the EuroFlow Consortium which received support from the FP6-2004-LIFESCIHEALTH-5 program of the European Commission (grant LSHB-CT-2006-018708) as Specific Targeted Research Project (STREP). The EuroFlow Consortium is part of the European Scientific Foundation for Hemato-Oncology (ESLHO), a Scientific Working Group (SWG) of the European Hematology Association (EHA); the grant of the Polish National Center for Research and Development (no. STRATEGMED3/304586/5/NCBR/2017 Person ALL); and internal grant of the Medical University of Silesia (no. PCN-1-050/K/0/K); the grant of CIBER-ONC, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad, Madrid, Spain and FONDOS FEDER (no. CB16/12/00400).
- Published
- 2022
- Full Text
- View/download PDF
12. Flow cytometric minimal residual disease assessment in B-cell precursor acute lymphoblastic leukaemia patients treated with CD19-targeted therapies
- Author
-
Martijn W. C. Verbeek, Chiara Buracchi, Anna Laqua, Stefan Nierkens, Lukasz Sedek, Juan Flores‐Montero, Mattias Hofmans, Elaine Sobral de Costa, Michaela Nováková, Ester Mejstrikova, Susana Barrena, Saskia Kohlscheen, Monika Szczepanowski, Jan Kulis, Elen Oliveira, Romana Jugooa, Anja X. Jong, Tomasz Szczepanski, Jan Philippé, Jacques J. M. Dongen, Alberto Orfao, Monika Brüggemann, Giuseppe Gaipa, Vincent H. J. Velden, Immunology, European Commission, European Hematology Association, Silesian University of Technology, Verbeek, M, Buracchi, C, Laqua, A, Nierkens, S, Sedek, L, Flores-Montero, J, Hofmans, M, Sobral de Costa, E, Novakova, M, Mejstrikova, E, Barrena, S, Kohlscheen, S, Szczepanowski, M, Kulis, J, Oliveira, E, Jugooa, R, de Jong, A, Szczepanski, T, Philippe, J, van Dongen, J, Orfao, A, Bruggemann, M, Gaipa, G, and van der Velden, V
- Subjects
acute leukaemia ,Neoplasm, Residual ,Minimal residual disease ,flow cytometry ,Antigens, CD19 ,hemic and immune systems ,Hematology ,diagnostic haematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Burkitt Lymphoma ,Acute leukaemia ,body regions ,Diagnostic haematology ,hemic and lymphatic diseases ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Medicine and Health Sciences ,minimal residual disease ,Humans ,Flow cytometry ,Adaptor Proteins, Signal Transducing - Abstract
The standardized EuroFlow protocol, including CD19 as primary B-cell marker, enables highly sensitive and reliable minimal residual disease (MRD) assessment in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) patients treated with chemotherapy. We developed and validated an alternative gating strategy allowing reliable MRD analysis in BCP-ALL patients treated with CD19-targeting therapies. Concordant data were obtained in 92% of targeted therapy patients who remained CD19-positive, whereas this was 81% in patients that became (partially) CD19-negative. Nevertheless, in both groups median MRD values showed excellent correlation with the original MRD data, indicating that, despite higher interlaboratory variation, the overall MRD analysis was correct., The EuroFlow Consortium received support from the FP6-2004-LIFESCIHEALTH-5 programme of the European Commission (grant LSHB-CT-2006-018708) as Specific Targeted Research Project (STREP). The EuroFlow Consortium is part of the European Scientific Foundation for Hemato-Oncology (ESLHO), a Scientific Working Group (SWG) of the European Hematology Association (EHA). TS and LS were supported by a Scientific Grant from the Medical University of Silesia Nr. PCN-1-050/K/0/K.
- Published
- 2021
13. Therapeutic afucosylated monoclonal antibody and bispecific T-cell engagers for T-cell acute lymphoblastic leukemia
- Author
-
Pierpaolo Correale, Andrea Ghelli Luserna di Rorà, Francesco Conforti, Gaetanina Golino, Caterina Riillo, Andrea Biondi, Massimo Martino, Giada Juli, Francesca Scionti, Pierfrancesco Tassone, Daniele Caracciolo, Emanuela Altomare, Gabriella Talarico, Eugénie Duroyon, Beatrice Belmonte, Nicoletta Polerà, Ludovic Lhermitte, Chiara Buracchi, Marco Rossi, Vahid Asnafi, Andrea Ballerini, Katia Grillone, Simona Sestito, Markus Hildinger, Greta Alampi, Cirino Botta, Maria Eugenia Gallo Cantafio, Maria Teresa Di Martino, Anna Maria Ferrari, Antonio Giordano, Mariamena Arbitrio, Pierosandro Tagliaferri, Licia Pensabene, Alessandro Gulino, Daniela Concolino, Giovanni Martinelli, Michelangelo Iannone, Claudio Tripodo, Giuseppe Gaipa, Caracciolo, Daniele, Riillo, Caterina, Ballerini, Andrea, Gaipa, Giuseppe, Lhermitte, Ludovic, Rossi, Marco, Botta, Cirino, Duroyon, Eugénie, Grillone, Katia, Gallo Cantafio, Maria Eugenia, Buracchi, Chiara, Alampi, Greta, Gulino, Alessandro, Belmonte, Beatrice, Conforti, Francesco, Golino, Gaetanina, Juli, Giada, Altomare, Emanuela, Polerà, Nicoletta, Scionti, Francesca, Arbitrio, Mariamena, Iannone, Michelangelo, Martino, Massimo, Correale, Pierpaolo, Talarico, Gabriella, Ghelli Luserna di Rorà, Andrea, Ferrari, Anna, Concolino, Daniela, Sestito, Simona, Pensabene, Licia, Giordano, Antonio, Hildinger, Marku, Di Martino, Maria Teresa, Martinelli, Giovanni, Tripodo, Claudio, Asnafi, Vahid, Biondi, Andrea, Tagliaferri, Pierosandro, Tassone, Pierfrancesco, Caracciolo, D, Riillo, C, Ballerini, A, Gaipa, G, Lhermitte, L, Rossi, M, Botta, C, Duroyon, E, Grillone, K, Cantafio, M, Buracchi, C, Alampi, G, Gulino, A, Belmonte, B, Conforti, F, Golino, G, Juli, G, Altomare, E, Polera, N, Scionti, F, Arbitrio, M, Iannone, M, Martino, M, Correale, P, Talarico, G, Di Rora, A, Ferrari, A, Concolino, D, Sestito, S, Pensabene, L, Giordano, A, Hildinger, M, Di Martino, M, Martinelli, G, Tripodo, C, Asnafi, V, Biondi, A, Tagliaferri, P, and Tassone, P
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,T-Lymphocytes ,Mice, SCID ,afucosylated monoclonal antibody ,Lymphocyte Activation ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Epitopes ,Jurkat Cells ,Antineoplastic Agents, Immunological ,Antibody Specificity ,Mice, Inbred NOD ,antigens ,Antibodies, Bispecific ,Tumor Microenvironment ,Immunology and Allergy ,antibodies ,hematologic neoplasms ,RC254-282 ,Antibody-dependent cell-mediated cytotoxicity ,Leukosialin ,bispecific T-cell engagers ,medicine.diagnostic_test ,biology ,hematological malignancie ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.anatomical_structure ,antibodie ,Oncology ,translational medical research ,Molecular Medicine ,Immunohistochemistry ,Female ,immunotherapy ,Antibody ,T-ALL ,T-cell engagers ,T-cell acute lymphoblastic leukemia ,medicine.drug_class ,T cell ,Immunology ,Settore MED/08 - Anatomia Patologica ,Monoclonal antibody ,Antibodies, Monoclonal, Humanized ,Flow cytometry ,T Acute Lymphoblastic Leukemia ,antigen ,Antigen ,Phagocytosis ,medicine ,Animals ,Humans ,hematological malignancies ,Cell Proliferation ,Pharmacology ,T-cell engager ,business.industry ,neoplasm ,translational research ,Basic Tumor Immunology ,Xenograft Model Antitumor Assays ,Cancer research ,biology.protein ,business ,hematologic neoplasm - Abstract
BackgroundT-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease with a poor cure rate for relapsed/resistant patients. Due to the lack of T-cell restricted targetable antigens, effective immune-therapeutics are not presently available and the treatment of chemo-refractory T-ALL is still an unmet clinical need. To develop novel immune-therapy for T-ALL, we generated an afucosylated monoclonal antibody (mAb) (ahuUMG1) and two different bispecific T-cell engagers (BTCEs) against UMG1, a unique CD43-epitope highly and selectively expressed by T-ALL cells from pediatric and adult patients.MethodsUMG1 expression was assessed by immunohistochemistry (IHC) on a wide panel of normal tissue microarrays (TMAs), and by flow cytometry on healthy peripheral blood/bone marrow-derived cells, on 10 different T-ALL cell lines, and on 110 T-ALL primary patient-derived cells. CD43-UMG1 binding site was defined through a peptide microarray scanning. ahuUMG1 was generated by Genetic Glyco-Engineering technology from a novel humanized mAb directed against UMG1 (huUMG1). BTCEs were generated as IgG1-(scFv)2 constructs with bivalent (2+2) or monovalent (2+1) CD3ε arms. Antibody dependent cellular cytotoxicity (ADCC), antibody dependent cellular phagocytosis (ADCP) and redirected T-cell cytotoxicity assays were analysed by flow cytometry. In vivo antitumor activity of ahUMG1 and UMG1-BTCEs was investigated in NSG mice against subcutaneous and orthotopic xenografts of human T-ALL.ResultsAmong 110 T-ALL patient-derived samples, 53 (48.1%) stained positive (24% of TI/TII, 82% of TIII and 42.8% of TIV). Importantly, no expression of UMG1-epitope was found in normal tissues/cells, excluding cortical thymocytes and a minority (2 log lower as compared with ahuUMG1, with significant mice survival advantage in different T-ALL models in vivo.ConclusionAltogether our findings, including the safe UMG1-epitope expression profile, provide a framework for the clinical development of these innovative immune-therapeutics for this still orphan disease.
- Published
- 2021
- Full Text
- View/download PDF
14. Automated identification of leukocyte subsets improves standardization of database-guided expert-supervised diagnostic orientation in acute leukemia: a EuroFlow study
- Author
-
Ludovic Lhermitte, Sylvain Barreau, Daniela Morf, Paula Fernandez, Georgiana Grigore, Susana Barrena, Maaike de Bie, Juan Flores-Montero, Monika Brüggemann, Ester Mejstrikova, Stefan Nierkens, Leire Burgos, Joana Caetano, Giuseppe Gaipa, Chiara Buracchi, Elaine Sobral da Costa, Lukasz Sedek, Tomasz Szczepański, Carmen-Mariana Aanei, Alita van der Sluijs-Gelling, Alejandro Hernández Delgado, Rafael Fluxa, Quentin Lecrevisse, Carlos E. Pedreira, Jacques J.M. van Dongen, Alberto Orfao, Vincent H.J. van der Velden, J. J.M. van Dongen, W.M. Bitter, B.R. Lubbers, C.I. Teodosio, M. Zlei, A.J. van der Sluijs-Gelling, F. de Bie, S. de Bruin-Versteeg, M. van der Burg, M.W. Schilham, V. H.J. van der Velden, A.W. Langerak, J. te Marvelde, A.E. Bras, J. Schilperoord-Vermeulen, R. Jugooa, K.C. Heezen, A. Orfao, J. Almeida, M.B. Vidriales, J. Flores-Montero, M. Pérez-Andrés, S. Matarraz, L. Martín, Q. Lecrevisse, J.J. Pérez-Morán, N. Puig, A. Medina Almeida, M. Gomes da Silva, T. Faria, M. Brüggemann, M. Ritgen, M. Szczepanowski, S. Kohlscheen, A. Laqua, E. Harbst, J. Finke, V. Asnafi, L. Lhermitte, E. Duroyon, J. Trka, O. Hrusak, T. Kalina, E. Mejstrikova, M. Novakova, D. Thurner, V. Kanderova, T. Szczepanski, L. Sędek, J. Bulsa, L. Slota, J. Kulis, C.E. Pedreira, E. Sobral da Costa, S. Nierkens, A. de Jong, A. de Koning, M. Lima, A.H. Santos, S. Böttcher, S. Lange, R. Engelmann, D. Paape, C. Machka, G. Gaipa, C. Burracchi, C. Bugarin, E. Lopez-Granados, L. del Pino Molina, L. Campos-Guyotat, C. Aanei, J. F. San Miguel, B. Paiva, L. Burgos, N. Villamor-Casas, L. Magnano, J. Philippé, C. Bonroy, B. Denys, A. Willems, P. Breughe, J. de Wolf, A.E. Sousa, S.L. Silva, P. Fernandez, D. Morf, European Commission, Instituto de Salud Carlos III, Ministerio de Economía y Competitividad (España), Agencia Estatal de Investigación (España), Ministerio de Ciencia, Innovación y Universidades (España), Silesian University of Technology, Lhermitte, L, Barreau, S, Morf, D, Fernandez, P, Grigore, G, Barrena, S, de Bie, M, Flores-Montero, J, Bruggemann, M, Mejstrikova, E, Nierkens, S, Burgos, L, Caetano, J, Gaipa, G, Buracchi, C, da Costa, E, Sedek, L, Szczepanski, T, Aanei, C, van der Sluijs-Gelling, A, Delgado, A, Fluxa, R, Lecrevisse, Q, Pedreira, C, van Dongen, J, Orfao, A, van der Velden, V, Bitter, W, Lubbers, B, Teodosio, C, Zlei, M, de Bie, F, de Bruin-Versteeg, S, van der Burg, M, Schilham, M, Langerak, A, te Marvelde, J, Bras, A, Schilperoord-Vermeulen, J, Jugooa, R, Heezen, K, Almeida, J, Vidriales, M, Perez-Andres, M, Matarraz, S, Martin, L, Perez-Moran, J, Puig, N, Almeida, A, Gomes da Silva, M, Faria, T, Ritgen, M, Szczepanowski, M, Kohlscheen, S, Laqua, A, Harbst, E, Finke, J, Asnafi, V, Duroyon, E, Trka, J, Hrusak, O, Kalina, T, Novakova, M, Thurner, D, Kanderova, V, Bulsa, J, Slota, L, Kulis, J, de Jong, A, de Koning, A, Lima, M, Santos, A, Bottcher, S, Lange, S, Engelmann, R, Paape, D, Machka, C, Burracchi, C, Bugarin, C, Lopez-Granados, E, del Pino Molina, L, Campos-Guyotat, L, Miguel, J, Paiva, B, Villamor-Casas, N, Magnano, L, Philippe, J, Bonroy, C, Denys, B, Willems, A, Breughe, P, de Wolf, J, Sousa, A, Silva, S, and Immunology
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Standardization ,Computer science ,Leukaemia, Laboratory techniques and procedures ,Article ,Immunophenotyping ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,EuroFlow ,Leukocytes ,medicine ,Humans ,Leukaemia ,Flow cytometry ,Future ,Acute leukemia ,Orientation (computer vision) ,business.industry ,Laboratory techniques and procedures ,Pattern recognition ,Flow Cytometry ,Peripheral blood ,Leukemia, Myeloid, Acute ,Identification (information) ,030104 developmental biology ,Área de Biomedicina ,T cell subset ,Artificial intelligence ,business ,Algorithms ,030215 immunology - Abstract
© The Author(s) 2020., Precise classification of acute leukemia (AL) is crucial for adequate treatment. EuroFlow has previously designed an AL orientation tube (ALOT) to guide toward the relevant classification panel and final diagnosis. In this study, we designed and validated an algorithm for automated (database-supported) gating and identification (AGI tool) of cell subsets within samples stained with ALOT. A reference database of normal peripheral blood (PB, n = 41) and bone marrow (BM; n = 45) samples analyzed with the ALOT was constructed, and served as a reference for the AGI tool to automatically identify normal cells. Populations not unequivocally identified as normal cells were labeled as checks and were classified by an expert. Additional normal BM (n = 25) and PB (n = 43) and leukemic samples (n = 109), analyzed in parallel by experts and the AGI tool, were used to evaluate the AGI tool. Analysis of normal PB and BM samples showed low percentages of checks ( 0.95 for all cell types in PB and r2 > 0.75 in BM) and resulted in highly concordant classification of leukemic cells by our previously published automated database-guided expert-supervised orientation tool for immunophenotypic diagnosis and classification of acute leukemia (Compass tool). Similar data were obtained using alternative, commercially available tubes, confirming the robustness of the developed tools. The AGI tool represents an innovative step in minimizing human intervention and requirements in expertise, toward a “sample-in and result-out” approach which may result in more objective and reproducible data analysis and diagnostics. The AGI tool may improve quality of immunophenotyping in individual laboratories, since high percentages of checks in normal samples are an alert on the quality of the internal procedures., The EuroFlow Consortium received support from the FP6- 2004-LIFESCIHEALTH-5 program of the European Commission (grant LSHB-CT-2006-018708) as Specific Targeted Research Project (STREP). The Prague team received support from the grant number NV18-03-00343. The Salamanca team received support from the Instituto de Salud Carlos III (ISCIII) (PI16/00787-FEDER) and from Agencia Estatal de Investigación (RTC-2016-4865-1-FEDER), Ministerio de Economía y Competitividad, Madrid, Spain. AHD is supported from the program DI-17-09591 from Agencia Estatal de Investigación, Ministerio de Ciencia, Innovación y Universidades, Madrid, Spain. SB is supported from the program PTQ16-08364 from Agencia Estatal de Investigación, Ministerio de Ciencia, Innovación y Universidades, Madrid, Spain. Medical University of Silesia in Katowice team was supported by the Strategmed III PersonALL grant [No. 304586/5/NCBR/2017] from the Polish National Center of Research and Development. The EuroFlow Consortium is part of the European Scientific Foundation for Hemato-Oncology (ESLHO), a Scientific Working Group (SWG) of the European Hematology Association (EHA).
- Published
- 2021
15. Monocyte-macrophage polarization and recruitment pathways in the tumour microenvironment of B-cell acute lymphoblastic leukaemia
- Author
-
Alessandra Fallati, Oscar Maglia, Giovanna D'Amico, Fabio Pagni, Mariella D'Angiò, Erica Dander, Federica Portale, Giulia Cricrì, Barbara Bottazzi, Fabio Pasqualini, Rita Starace, Lisa Brizzolara, A Mantovani, Chiara Buracchi, Paola Allavena, Gloria Bedini, Maria Grazia Valsecchi, Stefania Gaspari, Franco Locatelli, Andrea Biondi, Tamara Gulic, Daniela Silvestri, Cecilia Garlanda, Dander, E, Fallati, A, Gulic, T, Pagni, F, Gaspari, S, Silvestri, D, Cricri, G, Bedini, G, Portale, F, Buracchi, C, Starace, R, Pasqualini, F, D'Angio, M, Brizzolara, L, Maglia, O, Mantovani, A, Garlanda, C, Valsecchi, M, Locatelli, F, Biondi, A, Bottazzi, B, Allavena, P, and D'Amico, G
- Subjects
Adult ,Male ,Chemokine ,acute lymphoblastic leukaemia ,Adolescent ,CD14 ,chemokines ,macrophage ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,medicine ,Human Umbilical Vein Endothelial Cells ,Tumor Microenvironment ,Macrophage ,Humans ,CX3CL1 ,Aged ,bone marrow microenvironment ,biology ,Chemistry ,Monocyte ,Macrophages ,Mesenchymal stem cell ,chemokine ,Hematology ,Middle Aged ,Coculture Techniques ,Neoplasm Proteins ,medicine.anatomical_structure ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,030220 oncology & carcinogenesis ,mesenchymal stromal cell ,monocyte ,Cancer research ,biology.protein ,Female ,Bone marrow ,mesenchymal stromal cells ,CD163 ,030215 immunology - Abstract
B-cell acute lymphoblastic leukaemia (B-ALL) reprograms the surrounding bone marrow (BM) stroma to create a leukaemia-supportive niche. To elucidate the contribution of immune cells to the leukaemic microenvironment, we investigated the involvement of monocyte/macrophage compartments, as well as several recruitment pathways in B-ALL development. Immunohistochemistry analyses showed that CD68-expressing macrophages were increased in leukaemic BM biopsies, compared to controls and predominantly expressed the M2-like markers CD163 and CD206. Furthermore, the "non-classical" CD14+ CD16++ monocyte subset, expressing high CX3CR1 levels, was significantly increased in B-ALL patients' peripheral blood. CX3CL1 was shown to be significantly upregulated in leukaemic BM plasma, thus providing an altered migratory pathway possibly guiding NC monocyte recruitment into the BM. Additionally, the monocyte/macrophage chemoattractant chemokine ligand 2 (CCL2) strongly increased in leukaemic BM plasma, possibly because of the interaction of leukaemic cells with mesenchymal stromal cells and vascular cells and due to a stimulatory effect of leukaemia-related inflammatory mediators. C5a, a macrophage chemoattractant and M2-polarizing factor, further appeared to be upregulated in the leukaemic BM, possibly as an effect of PTX3 decrease, that could unleash complement cascade activation. Overall, deregulated monocyte/macrophage compartments are part of the extensive BM microenvironment remodelling at B-ALL diagnosis and could represent valuable targets for novel treatments to be coupled with classical chemotherapy.
- Published
- 2020
16. Flow cytometry for minimal residual disease testing in acute leukemia: opportunities and challenges
- Author
-
Giuseppe Gaipa, Chiara Buracchi, Andrea Biondi, Gaipa, G, Buracchi, C, and Biondi, A
- Subjects
Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,Myeloid ,Immunophenotyping ,Pathology and Forensic Medicine ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Genetics ,medicine ,Humans ,Neoplasm ,Molecular Biology ,Acute leukemia ,medicine.diagnostic_test ,business.industry ,flow cytometry ,medicine.disease ,Molecular medicine ,Minimal residual disease ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Acute Disease ,minimal residual disease ,Molecular Medicine ,Acute Leukemia ,business ,030215 immunology - Abstract
Introduction: Flow cytometric quantification of minimal residual disease (MRD) in acute leukemia (AL) represents an indispensable tool to guide modern therapeutic protocols toward a precision medicine approach, being a powerful predictor of the overall response to treatment. This review covers the most challenging aspects and developments of this method, aiming at supporting further its implementation in clinical practices. Area covered: Flow cytometric MRD is based on the discrimination of leukemia cells from their physiological counterparts by the recognition of the leukemia-associated immunophenotypes. Technical and standardization advances along the last decades have been implemented allowing flow cytometric MRD to consolidate its role in modern therapeutic protocols for ALs. However, gaps in sensitivity and data interpretation are still present together with the need for further optimization of MRD-based clinical protocols. In this review, we critically analyze and discuss the most relevant and representative contributions in the field by accurate selection of the literature available in PubMed. Expert commentary: Further research in flow cytometric MRD can bring this technology toward wider and consistent applications in multiple acute leukemia settings rendering this tool a future golden standard and providing clinicians with more reliable and accurate tools for clinical decisions.
- Published
- 2018
- Full Text
- View/download PDF
17. Donor-Derived CAR T Cells Engineered with Sleeping Beauty in Pediatric and Adult Patients with Acute Lymphoblastic Leukemia Relapsed Post-HSCT
- Author
-
Giovanni Cazzaniga, Chiara F. Magnani, Gianluca Cavallaro, Giuseppe Gaipa, Giuseppe Dastoli, Gian Maria Borleri, Andrea Biondi, Sarah Tettamanti, Silvia Ferrari, Giuliana Rizzuto, Adriana Balduzzi, Federico Lussana, Maria Grazia Valsecchi, Giuseppe Gritti, Benedetta Rambaldi, Alessandro Rambaldi, Chiara Buracchi, Martino Introna, Sara Napolitano, Daniela Belotti, Silvia Zaninelli, and Stefania Galimberti
- Subjects
Oncology ,medicine.medical_specialty ,Adult patients ,business.industry ,Lymphoblastic Leukemia ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Internal medicine ,medicine ,Donor derived ,Car t cells ,business - Abstract
Introduction Allogeneic Chimeric Antigen Receptor (CAR) T cells engineered with non-viral methods offer a modality to reduce costs and logistical complexity of the viral process and allow lymphodepleted patients to access CAR T cell treatment. We recently proposed the use of Sleeping Beauty (SB) transposon to engineer donor-derived T cells differentiated according to the cytokine-induced killer (CIK) cell protocol (Magnani CF et al. J Clin Invest. 2021). We report here outcomes on B-cell acute lymphoblastic leukemia (B-ALL) patients, relapsing after transplantation, treated with donor-derived anti-CD19 CAR T cells (CARCIK-CD19). Methods We conducted an academic, multi-center, phase I/II dose-escalation trial in patients relapsed after allogeneic hematopoietic stem cell transplantation (HSCT). The infusion product was manufactured in-house starting from 50 mL of peripheral blood from the HSCT donor by electroporation with GMP-grade plasmids. All patients underwent lymphodepletion with Fludarabine (30 mg/m 2/day x 4 days) and Cyclophosphamide (500 mg/m 2/day x 2 days), before proceeding to CARCIK-CD19 infusion. We used the Bayesian Optimal Interval (BOIN) design to define a four-dose escalation scheme. Primary objectives were to define the Maximum Tolerated Dose (MTD), safety, and feasibility. Secondary objectives included the assessment of complete hematologic response (CR), duration of response (DOR), progression-free (PFS), event-free (EFS), and overall survival (OS). This study was registered at ClinicalTrials.gov, NCT03389035. Results From January 2018 to June 2021, a total of 32 patients were screened, 26 enrolled (6 children and 20 adults) and 21 infused (4 children and 17 adults). Reasons for not receiving infusion included consent withdrawal (N=1), disease progression not controlled by bridging therapy (N=3), acquisition of myeloid phenotype (N=1). The median number of prior therapies was 4 (range, 1-7) with a median time interval from HSCT to relapse of 9 months. The median BM blasts was 60% (range, 5-100%) at enrollment and 7% (range, 0-96%) post lymphodepletion. Of the 21 patients infused, CARCIK-CD19 were obtained by HLA-identical sibling (n=6, 29%), matched unrelated (n= 7, 33%), and haploidentical donors (n=8, 38%). Three patients (14%) received the first dose level of 1x10 6 CARCIK-CD19 cells/Kg, three (14%) the second of 3x10 6, and three (14%) the third of 7.5x10 6 whereas 12 patients (57%) received the fourth and last planned dose level of 15x10 6 cells/Kg, as no dose limiting toxicity (DLT) was observed. CRS was observed in six patients (three grade I and three grade II) and immune effector cell-associated neurotoxicity in two patients at the highest dose. Although 9 out of 21 had experienced acute or chronic graft-versus-host disease (GvHD) after the previous HSCT, secondary GvHD was never induced by CARCIK-CD19. Complete response was achieved by 13 out of 21 patients (61.9%, 95%CI=38-82%) and by 11 out of 15 patients treated with the 2 highest doses (73.3%, 95%CI=45-92%). Eleven of these responders were MRD-negative. Notably, the type of donor did not influence the achievement of CR 28 days post-infusion. At a median follow up of 21.6 months (range, 1.0-38.4 months), 10 patients (47.6%) are alive in CR (9 in the 2 highest dose levels). Overall, the median OS and EFS were 9.7 and 3.2 months, respectively, with a median DOR of 4.0 months (range, 1.0-23.5 months). Patients in CR at 28-days had a 6-months relapse-free survival of 48.4% (SE=14.9). EFS at 6 months was 26.5% (SE=9.9) and OS was 67.6% (SE=11.1). Among the 13 patients who achieved CR, two children underwent consolidation with a second allo-HSCT in complete remission. Adult patients did not receive any additional anti-leukemic therapies unless a relapse occurred, and four of them remained in remission and alive (+24, +9, +6, and +4 months). Robust CARCIK-CD19 cell expansion was achieved in most patients and CARCIK-CD19 cells were measurable for up to 22 months. Conclusions SB-engineered CAR T cells induce sustained responses in B-ALL patients relapsed after HSCT irrespective of the donor type and without severe toxicities. Disclosures Lussana: Incyte: Honoraria; Pfizer: Honoraria; Astellas Pharma: Honoraria; Amgen: Honoraria. Gritti: Takeda: Consultancy; Roche: Consultancy; Kite Gilead: Consultancy; IQvia: Consultancy; Italfarmaco: Consultancy; Clinigen: Consultancy. Biondi: Incyte: Consultancy, Other: Advisory Board; Bluebird: Other: Advisory Board; Novartis: Honoraria; Amgen: Honoraria; Colmmune: Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
18. Targeting CD33 in Chemoresistant AML Patient-Derived Xenografts by CAR-CIK Cells Modified with an Improved SB Transposon System
- Author
-
Chiara F. Magnani, Marta Serafini, Maria Grazia Valsecchi, Tamás Raskó, Vincenzo Perriello, Ettore Biagi, Sarah Tettamanti, Gaia Alberti, Martino Introna, Zsuzsanna Izsvák, Maria Caterina Rotiroti, Felix Lundberg, Giuseppe Dastoli, Claudia Cappuzzello, Silvia Arcangeli, Chiara Buracchi, Amit Pande, Andrea Biondi, Stefania Galimberti, Rotiroti, M, Buracchi, C, Arcangeli, S, Galimberti, S, Valsecchi, M, Perriello, V, Rasko, T, Alberti, G, Magnani, C, Cappuzzello, C, Lundberg, F, Pande, A, Dastoli, G, Introna, M, Serafini, M, Biagi, E, Izsvák, Z, Biondi, A, and Tettamanti, S
- Subjects
Myeloid ,Cell Transplantation ,THP-1 Cells ,medicine.medical_treatment ,CD33 ,Sialic Acid Binding Ig-like Lectin 3 ,Adoptive ,Drug Resistance ,Transposases ,Mice, SCID ,Immunotherapy, Adoptive ,Cell therapy ,Mice ,0302 clinical medicine ,AML ,Mice, Inbred NOD ,Drug Discovery ,Receptors ,Medicine ,Cell Engineering ,0303 health sciences ,Receptors, Chimeric Antigen ,Leukemia ,Cytokine-induced killer cell ,Gene Transfer Techniques ,CAR ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Treatment Outcome ,030220 oncology & carcinogenesis ,Molecular Medicine ,Heterografts ,Original Article ,Immunotherapy ,cytokine-induced killer cells ,Context (language use) ,Acute ,SCID ,03 medical and health sciences ,Experimental ,Genetics ,Animals ,Humans ,Molecular Biology ,B cell ,030304 developmental biology ,Pharmacology ,Leukemia, Experimental ,business.industry ,Chimeric Antigen ,Genetic Therapy ,Sleeping Beauty transposon system ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,Drug Resistance, Neoplasm ,non-viral gene transfer ,Cancer research ,Sleeping Beauty transposon ,Feasibility Studies ,Inbred NOD ,Neoplasm ,business ,cytokine-induced killer cell - Abstract
The successful implementation of chimeric antigen receptor (CAR)-T cell therapy in the clinical context of B cell malignancies has paved the way for further development in the more critical setting of acute myeloid leukemia (AML). Among the potentially targetable AML antigens, CD33 is insofar one of the main validated molecules. Here, we describe the feasibility of engineering cytokine-induced killer (CIK) cells with a CD33.CAR by using the latest optimized version of the non-viral Sleeping Beauty (SB) transposon system "SB100X-pT4." This offers the advantage of improving CAR expression on CIK cells, while reducing the amount of DNA transposase as compared to the previously employed "SB11-pT" version. SB-modified CD33.CAR-CIK cells exhibited significant antileukemic activity invitro and invivo in patient-derived AML xenograft models, reducing AML development when administered as an "early treatment" and delaying AML progression in mice with established disease. Notably, by exploiting an already optimized xenograft chemotherapy model that mimics human induction therapy in mice, we demonstrated for the first time that CD33.CAR-CIK cells are also effective toward chemotherapy resistant/residual AML cells, further supporting its future clinical development and implementation within the current standard regimens.
- Published
- 2020
19. Standardized flow cytometry for highly sensitive MRD measurements in B-cell acute lymphoblastic leukemia
- Author
-
Prisca M J Theunissen, Edwin Sonneveld, Lukasz Sedek, Giuseppe Gaipa, Ludovic Lhermitte, Monika Brüggemann, Elaine Sobral da Costa, Vincent H.J. van der Velden, Alberto Orfao, Ester Mejstrikova, Jan Trka, Alita J. van der Sluijs-Gelling, Quentin Lecrevisse, Georgiana Grigore, Jeroen G. te Marvelde, Paola Bonaccorso, Tomasz Szczepański, Eva Froňková, Chiara Buracchi, Marius Bartels, Ondrej Hrusak, Michaela Novakova, Elen Oliveira, Michaela Kotrova, Jacques J.M. van Dongen, Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (Brasil), Ministerio de Educación, Cultura y Deporte (España), Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro, European Commission, Ministry of Health of the Czech Republic, Conselho Nacional de Desenvolvimento Científico e Tecnológico (Brasil), Czech Science Foundation, Immunology, Theunissen, P, Mejstrikova, E, Sedek, L, Van Der Sluijs-Gelling, A, Gaipa, G, Bartels, M, Sobral da Costa, E, Kotrova, M, Novakova, M, Sonneveld, E, Buracchi, C, Bonaccorso, P, Oliveira, E, Te Marvelde, J, Szczepanski, T, Lhermitte, L, Hrusak, O, Lecrevisse, Q, Grigore, G, Fronkova, E, Trka, J, Bruggemann, M, Orfao, A, Van Dongen, J, and Van Der Velden, V
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Neoplasm, Residual ,Adolescent ,Immunology ,MRD, flow cytometry, B-cell acute lymphoblastic leukemia ,Receptors, Antigen, B-Cell ,Biology ,Real-Time Polymerase Chain Reaction ,Sensitivity and Specificity ,Biochemistry ,Flow cytometry ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,EuroFlow ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Acute lymphocytic leukemia ,medicine ,Humans ,Child ,Aged ,Gene Rearrangement ,medicine.diagnostic_test ,Infant, Newborn ,Infant ,Cell Biology ,Hematology ,Gene rearrangement ,Middle Aged ,Flow Cytometry ,medicine.disease ,Minimal residual disease ,medicine.anatomical_structure ,Child, Preschool ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Bone marrow ,Antibody ,030215 immunology - Abstract
A fully-standardized EuroFlow 8–color antibody panel and laboratory procedure was stepwise designed to measure minimal residual disease (MRD) in B-cell precursor (BCP) acute lymphoblastic leukemia (ALL) patients with a sensitivity of £10, comparable to real-time quantitative polymerase chain reaction (RQ-PCR)–based MRD detection via antigen-receptor rearrangements. Leukocyte markers and the corresponding antibodies and fluorochromes were selected based on their contribution in separating BCP-ALL cells from normal/regenerating BCP cells in multidimensional principal component analyses. After 5 multicenter design-test-evaluate-redesign phases with a total of 319 BCP-ALL patients at diagnosis, two 8-color antibody tubes were selected, which allowed separation between normal and malignant BCP cells in 99% of studied patients. These 2 tubes were tested with a new erythrocyte bulk-lysis protocol allowing acquisition of high cell numbers in 377 bone marrow follow-up samples of 178 BCP-ALL patients. Comparison with RQ-PCR–based MRD data showed a clear positive relation between the percentage concordant cases and the number of cells acquired. For those samples with >4 million cells acquired, concordant results were obtained in 93% of samples. Most discordances were clarified upon high-throughput sequencing of antigen-receptor rearrangements and blind multicenter reanalysis of flow cytometric data, resulting in an unprecedented concordance of 98% (97% for samples with MRD < 0.01%). In conclusion, the fully standardized EuroFlow BCP-ALL MRD strategy is applicable in >98% of patients with sensitivities at least similar to RQ-PCR (£10), if sufficient cells (>4 3 10, preferably more) are evaluated., E.F. was supported by the Grant Agency of the Czech Republic (project of Centre of Excellence No. P302/12/G101). L.S., T.S., and P.T. were supported by European Research Area Network (ERA-NET) PrioMedChild, grant 40-41800-98-027. E.M. was supported by Ministry of Health of the Czech Republic, grant 15-28525A. M.K. was supported by the University Hospital Motol, Prague, Czech Republic (00064203). E.S.d.C., Q.L., and A.O. acknowledge the Bilateral Cooperation Program between Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (Brasília, Brazil) and Dirección General de Políticas Universitárias–Ministério de Educación, Cultura y Deportes (Madrid, Spain) (311/15). E.S.d.C. acknowledges Research Foundation of the State of Rio de Janeiro, Rio de Janeiro, Brazil (E26/110.105/2014, E26/102.191/2013) and Conselho Nacional de Desenvolvimento Científico e Tecnológico–CNPQ of Brazil (400194/2014-7). G.G., C.B., and P.B. were supported by Fondazione Tettamanti.
- Published
- 2017
- Full Text
- View/download PDF
20. Donor-Derived CAR T Cells Engineered with Sleeping Beauty Achieve Anti-Leukemic Activity without Severe Toxicity
- Author
-
Chiara F. Magnani, Silvia Ferrari, Giuliana Rizzuto, Sarah Tettamanti, Federico Lussana, Fabrizio Benedicenti, Valentina Colombo, Ettore Biagi, Giovanni Cazzaniga, Attilio Rovelli, Stefania Galimberti, Silvia Zaninelli, Maria Grazia Valsecchi, Giuseppe Gritti, Benedetta Cabiati, Martino Introna, Andrea Biondi, Sara Napolitano, Gian Maria Borleri, Chiara Buracchi, Eugenio Montini, Daniela Belotti, Alessandro Rambaldi, Giuseppe Dastoli, Giuseppe Gaipa, and Adriana Balduzzi
- Subjects
medicine.medical_specialty ,education.field_of_study ,Cyclophosphamide ,business.industry ,medicine.medical_treatment ,T cell ,Immunology ,Population ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Fludarabine ,Cytokine release syndrome ,medicine.anatomical_structure ,Internal medicine ,Clinical endpoint ,Medicine ,business ,education ,CD8 ,medicine.drug - Abstract
Background Significant efforts over the past few years led Chimeric Antigen Receptor (CAR) T cell therapy to success in relapsed and refractory (r/r) B-cell malignancies. Still logistical complexity, high costs and toxicities are currently the main barriers to the use of CAR T cell therapy. We therefore propose non-viral engineering of an allogeneic T cell population according to cytokine induced killer (CIK) cell protocol of differentiation. Methods We reported the updated results of our phase I/II trial in B-cell acute lymphoblastic leukemia (B-ALL) patients relapsed after allogeneic hematopoietic stem cell transplantation (HSCT) using donor-derived CD19 CAR T cells generated with the Sleeping Beauty (SB) transposon and differentiated into CIK (CARCIK-CD19) according to the method enclosed in the filed patent EP20140192371. After lymphodepletion with Fludarabine (30 mg/m2/day) x 4 days and Cyclophosphamide (500 mg/m2/day) x 2 days, CARCIK-CD19 were infused following a four-dose escalation scheme (1x106, 3x106, 7.5x106 and 15x106 transduced CAR+ T cells/kg) according to the Bayesian Optimal Interval Design (BOIN). During the cell manufacturing period, bridging anti leukemic therapy from patient registration to the beginning of the lymphodepletion, was allowed. The primary endpoint was to define the Maximum Tolerated Dose (MTD) and the safety assessment. Key secondary endpoints included the assessment of complete hematologic response (CR), defined as < 5% bone marrow (BM) blasts, circulating blasts < 1%, no clinical evidence of extramedullary disease, as well as the characterization of CARCIK-CD19 persistence in PB and BM (NCT03389035). Results The cellular product was produced successfully for all patients starting from the donor-derived peripheral blood (PB) and consisted mostly of CD3+ lymphocytes (mean 98.85% ±SD 1.19%) with a mean of 38.6% CAR expression (range 15.10%-73.17%). From January 2018 to July 2020, a total of 24 patients were screened, and 15 were enrolled (4 children and 11 adults) and infused with a single dose of CARCIK-CD19 (n=3 HLA identical sibling, n=4 MUD, n=8 haploidentical donor). The leukemic burden in the BM post lymphodepletion/pre-infusion ranged from 0% to 96%. Robust expansion was achieved in the majority of the patients. The maximal expansion reached about 1x106 transgene copies per μg DNA and 70% of CAR+ T cells in PB. CD8+ T cells represented the predominant circulating CAR+ T cell subset. Persistence of central memory CAR+ T cells was observed after infusion and CAR T cells were measurable up to 9 months. CARCIK-CD19 were characterized by a high profile of safety in all treated patients. Toxicities reported were two grade I and two grade II cytokine release syndrome (CRS) cases at the highest dose in the absence of graft-versus-host disease (GvHD), neurotoxicity, or dose-limiting toxicities. Seven out of 9 patients, receiving the highest doses, achieved CR and CRi at day 28. MRD-negative status for all responders was achieved by 6 out of 9 patients (1 currently in evaluation). The two patients in CR but with MRD+ relapsed with a CD19+ disease at +2.3 and +1.9 months post infusion, respectively. Among the 6 patients who achieved MRD-negative CR, two children underwent consolidation with a second allo-HSCT and are still alive and disease free (+17 and +13 months), two adult patients died of subsequent CD19+ disease relapse and two adult patients are still alive and disease free (+14 and +12 months) without additional therapies. The distribution profile of integration sites (IS) showed no preference for gene dense or promoter regions, and no particular differences between pre- and post- infusion sample IS. Samples harvested at early time points after infusion showed a highly polyclonal repertoire. At later time points (≥ 28 days after infusion) the repertoire of IS showed a marked reduction towards oligoclonality, in absence of specific dominant clones. Conclusions We can conclude that SB-engineered CAR T cells expand and persist in pediatric and adult B-ALL patients relapsed after HSCT. Sustained response was achieved without severe toxicities. All analyzed samples appear to have a highly polyclonal IS repertoire and no signs of genotoxicity by transposon insertions could be observed. Disclosures Gritti: IQVIA: Consultancy; Amgen: Honoraria; Autolus: Consultancy; Italfarmaco: Consultancy; F. Hoffmann-La Roche Ltd: Honoraria; Jannsen: Other: Travel Support; Takeda: Honoraria; Kite: Consultancy. Rambaldi:Sanofi: Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Omeros: Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company). Research grant from Amgen Inc.; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company). Advisory board and speaker fees from Pfizer.; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel support from Gilead.; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Support of parent study and funding of editorial support. Received travel support., Research Funding; University of Milan: Current Employment; BMS/Celgene: Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company); Astellas: Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company).
- Published
- 2020
- Full Text
- View/download PDF
21. Differential expression of CD73, CD86 and CD304 in normal vs. leukemic B-cell precursors and their utility as stable minimal residual disease markers in childhood B-cell precursor acute lymphoblastic leukemia
- Author
-
Alita J. van der Sluijs-Gelling, Jacques J.M. van Dongen, Prisca M J Theunissen, Vincent H.J. van der Velden, Michaela Novakova, Łukasz Sędek, Alberto Orfao, Ester Mejstrikova, Chiara Buracchi, Tomasz Szczepański, Giuseppe Gaipa, Magdalena Twardoch, Elen Oliveira, Alicja Sonsala, Elaine Sobral da Costa, Government of Czech Republic, European Commission, Immunology, Sedek, L, Theunissen, P, Sobral da Costa, E, van der Sluijs-Gelling, A, Mejstrikova, E, Gaipa, G, Sonsala, A, Twardoch, M, Oliveira, E, Novakova, M, Buracchi, C, van Dongen, J, Orfao, A, van der Velden, V, and Szczepanski, T
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Neoplasm, Residual ,Immunology ,MathematicsofComputing_GENERAL ,Acute lymphoblastic leukemia ,GPI-Linked Proteins ,Gastroenterology ,Flow cytometry ,5'-nucleotidase ,Fusion gene ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Internal medicine ,Neuropilin 1 ,Biomarkers, Tumor ,medicine ,Humans ,Immunology and Allergy ,CD86 ,Child ,5'-Nucleotidase ,B cell ,medicine.diagnostic_test ,business.industry ,Precursor Cells, B-Lymphoid ,Minimal residual disease ,TheoryofComputation_GENERAL ,GPI-Linked Protein ,Neuropilin-1 ,030104 developmental biology ,medicine.anatomical_structure ,CD304 ,Child, Preschool ,030220 oncology & carcinogenesis ,CD73 ,ComputingMilieux_COMPUTERSANDSOCIETY ,Female ,B7-2 Antigen ,business ,Human - Abstract
On behalf of the EuroFlow Consortium., [Background]: Optimal discrimination between leukemic blasts and normal B-cell precursors (BCP) is critical for treatment monitoring in BCP acute lymphoblastic leukemia (ALL); thus identification of markers differentially expressed on normal BCP and leukemic blasts is required., [Methods]: Multicenter analysis of CD73, CD86 and CD304 expression levels was performed in 282 pediatric BCP-ALL patients vs. normal bone marrow BCP, using normalized median fluorescence intensity (nMFI) values., [Results]: CD73 was expressed at abnormally higher levels (vs. pooled normal BCP) at diagnosis in 71/108 BCP-ALL patients (66%), whereas CD304 and CD86 in 119/202 (59%) and 58/100 (58%) patients, respectively. Expression of CD304 was detected at similar percentages in common-ALL and pre-B-ALL, while found at significantly lower frequencies in pro-B-ALL. A significant association (p = 0.009) was found between CD304 expression and the presence of the ETV6-RUNX1 fusion gene. In contrast, CD304 showed an inverse association with MLL gene rearrangements (p = 0.01). The expression levels of CD73, CD86 and CD304 at day 15 after starting therapy (MRD15) were stable or higher than at diagnosis in 35/37 (95%), 40/56 (71%) and 19/41 (46%) cases investigated, respectively. This was also associated with an increased mean nMFI at MRD15 vs. diagnosis of +24 and +3 nMFI units for CD73 and CD86, respectively. In addition, gain of expression of CD73 and CD86 at MRD15 for cases that were originally negative for these markers at diagnosis was observed in 16% and 18% of cases, respectively. Of note, CD304 remained aberrantly positive in 63% of patients, despite its levels of expression decreased at follow-up in 54% of cases., [Conclusions]: Here we show that CD73, CD86 and CD304 are aberrantly (over)expressed in a substantial percentage of BCP-ALL patients and that their expression profile remains relatively stable early after starting therapy, supporting their potential contribution to improved MRD analysis by flow cytometry., EM5 and MN5 were supported by the Czech Republic national BCP-ALL MRD grant AZV, no. 15-28525A. ŁS1 and TS7 were supported by the grant from the National Center of Research and Development Strategmed III PersonALL (No. 304586/5/NCBR/2017), internal grant from the Medical University of Silesia and by Iskierka Foundation (Katowice, Poland). PT2, TS7 and ŁS1 were financially supported by ERA-NET PrioMedChild project (no. 40-41800-98-027). Part of this research was performed within the framework of the Erasmus Postgraduate School Molecular Medicine.
- Published
- 2019
22. The atypical chemokine receptor ACKR2 drives pulmonary fibrosis by tuning influx of CCR2
- Author
-
Remo C, Russo, Benedetta, Savino, Massimiliano, Mirolo, Chiara, Buracchi, Giovanni, Germano, Achille, Anselmo, Luca, Zammataro, Fabio, Pasqualini, Alberto, Mantovani, Massimo, Locati, and Mauro M, Teixeira
- Subjects
Mice, Knockout ,Bleomycin ,Interferon-gamma ,Mice ,Receptors, CCR5 ,Receptors, CCR2 ,Pulmonary Fibrosis ,Animals ,Th17 Cells ,Receptors, Antigen, T-Cell, gamma-delta ,Chemokines - Abstract
Chemokines coordinate lung inflammation and fibrosis by acting on chemokine receptors expressed on leukocytes and other cell types. Atypical chemokine receptors (ACKRs) bind, internalize, and degrade chemokines, tuning homeostasis and immune responses. ACKR2 recognizes and decreases the levels of inflammatory CC chemokines. The role of ACKR2 in fibrogenesis is unknown. The purpose of the study was to investigate the role of ACKR2 in the context of pulmonary fibrosis. The effects of ACKR2 expression and deficiency during inflammation and fibrosis were analyzed using a bleomycin-model of fibrosis, ACKR2-deficient mice, bone marrow chimeras, and antibody-mediated leukocyte depletion. ACKR2 was upregulated acutely in response to bleomycin and normalized over time. ACKR2
- Published
- 2018
23. The atypical chemokine receptor ACKR2 drives pulmonary fibrosis by tuning influx of CCR2+ and CCR5+ IFNγ-producing γδT cells in mice
- Author
-
Alberto Mantovani, Massimiliano Mirolo, Mauro M. Teixeira, Achille Anselmo, Fabio Pasqualini, Giovanni Germano, Chiara Buracchi, Benedetta Savino, Luca Zammataro, Remo Castro Russo, Massimo Locati, Russo, R, Savino, B, Mirolo, M, Buracchi, C, Germano, G, Anselmo, A, Zammataro, L, Pasqualini, F, Mantovani, A, Locati, M, and Teixeira, M
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,CCR2 ,Chemokine ,ACKR2 ,Physiology ,Knockout ,Pulmonary Fibrosis ,Inflammation ,Bleomycin ,γδT lymphocytes ,03 medical and health sciences ,chemistry.chemical_compound ,Chemokine receptor ,Interferon-γ ,Pulmonary fibrosis ,Animals ,Chemokines ,Interferon-gamma ,Mice ,Mice, Knockout ,Receptors, Antigen, T-Cell, gamma-delta ,Receptors, CCR2 ,Receptors, CCR5 ,Th17 Cells ,0302 clinical medicine ,Fibrosis ,Physiology (medical) ,Receptors ,medicine ,Interferon gamma ,γδT lymphocyte ,gamma-delta ,biology ,business.industry ,Cell Biology ,medicine.disease ,T-Cell ,030104 developmental biology ,chemistry ,Antigen ,biology.protein ,Cancer research ,medicine.symptom ,business ,Pulmonary fibrosi ,CCR5 ,030215 immunology ,medicine.drug - Abstract
Chemokines coordinate lung inflammation and fibrosis by acting on chemokine receptors expressed on leukocytes and other cell types. Atypical chemokine receptors (ACKRs) bind, internalize, and degrade chemokines, tuning homeostasis and immune responses. ACKR2 recognizes and decreases the levels of inflammatory CC chemokines. The role of ACKR2 in fibrogenesis is unknown. The purpose of the study was to investigate the role of ACKR2 in the context of pulmonary fibrosis. The effects of ACKR2 expression and deficiency during inflammation and fibrosis were analyzed using a bleomycin-model of fibrosis, ACKR2-deficient mice, bone marrow chimeras, and antibody-mediated leukocyte depletion. ACKR2 was upregulated acutely in response to bleomycin and normalized over time. ACKR2−/− mice showed reduced lethality and lung fibrosis. Bone marrow chimeras showed that lethality and fibrosis depended on ACKR2 expression in pulmonary resident (nonhematopoietic) cells but not on leukocytes. ACKR2−/− mice exhibited decreased expression of tissue-remodeling genes, reduced leukocyte influx, pulmonary injury, and dysfunction. ACKR2−/− mice had early increased levels of CCL5, CCL12, CCL17, and IFNγ and an increased number of CCR2+ and CCR5+ IFNγ-producing γδT cells in the airways counterbalanced by low Th17-lymphocyte influx. There was reduced accumulation of IFNγ-producing γδT cells in CCR2−/− and CCR5−/− mice. Moreover, depletion of γδT cells worsened the clinical symptoms induced by bleomycin and reversed the phenotype of ACKR2−/− mice exposed to bleomycin. ACKR2 controls the CC chemokine expression that drives the influx of CCR2+ and CCR5+ IFNγ-producing γδT cells, tuning the Th17 response that mediated pulmonary fibrosis triggered by bleomycin instillation.
- Published
- 2018
24. Donor-Derived CD19 CAR Cytokine Induced Killer (CIK) Cells Engineered with Sleeping Beauty Transposon for Relapsed B-Cell Acute Lymphoblastic Leukemia (B-ALL)
- Author
-
Sarah Tettamanti, Silvia Rigamonti, Chiara Buracchi, Fabrizio Benedicenti, Adriana Balduzzi, Giovanni Cazzaniga, Alessandro Rambaldi, Benedetta Cabiati, Silvia Ferrari, Attilio Rovelli, Giuseppe Gritti, Andrea Biondi, Giada Matera, Giuseppe Dastoli, Silvia Zaninelli, Grazia Fazio, Martino Introna, Gian Maria Borleri, Giuseppe Gaipa, Chiara F. Magnani, Sara Napolitano, Eugenio Montini, Federico Lussana, Stefania Cesana, Valentina Colombo, and Daniela Belotti
- Subjects
biology ,business.industry ,Genetic enhancement ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Sleeping Beauty transposon system ,Biochemistry ,CD19 ,Transplantation ,Cytokine release syndrome ,Cytokine ,Acute lymphocytic leukemia ,biology.protein ,medicine ,Cancer research ,business - Abstract
Background Immunotherapy using patient-derived CAR T cells has achieved complete remission and durable response in highly refractory populations. However, logistical complexity and high costs of manufacturing autologous viral products limit CAR T cell availability. Allogeneic Cytokine Induced Killer (CIK) cells, a T-cell population characterized by the enrichment of CD3+CD56+ cells, have demonstrated a high profile of safety in acute lymphoblastic leukemia (ALL) patients (Introna M et al. Biol Blood Marrow Transplant. 2017). CIK cells could be easily engineered by the non-viral Sleeping Beauty (SB) transposon for the clinical application (Magnani CF et al, Hum Gene Ther. 2018, Biondi A et al. J Autoimmun. 2017). Methods CIK cells were generated from 50 ml of donor-derived peripheral blood (PB) by electroporation with the GMP-grade CD19.CAR/pTMNDU3 and pCMV-SB11 plasmids according to the method enclosed in the filed patent EP20140192371. After lymphodepletion with Fludarabine (30 mg/m2/day) x 4 days and Cyclophosphamide (300 mg/m2/day) x 2 days, CARCIK-CD19 were infused in pediatric and adult B-cell ALL (B-ALL) patients relapsed after allogeneic hematopoietic stem cell transplantation (HSCT). The clinical trial follows a four-dose escalation scheme (1x106, 3x106, 7.5x106 and 15x106 transduced CAR+ T cells/kg) using the novel Bayesian Optimal Interval Design (BOIN). During the cell manufacturing period, bridging anti leukemic therapy from patient registration to the beginning of the lymphodepletion, was allowed. The primary endpoint was to define the Maximum Tolerated Dose (MTD) and a safety assessment. Key secondary endpoints included the assessment of complete hematologic response (CR), defined as < 5% bone marrow (BM) blasts, circulating blasts < 1%, no clinical evidence of extramedullary disease, as well as the characterization of CARCIK-CD19 persistence in PB and BM (NCT03389035). Results We manufactured eighteen batches by seeding a median of 126.8x106 allogeneicPBMCs. At the end of expansion, the mean harvesting was 6.46x109 nucleated cells (range 1.39 - 16.00x109). Manufactured cells were mostly CD3+ lymphocytes (mean 98.90% ±SE 0.30%). Of these, 43.57%±3.73% were CAR+, 47.07%±2.74% were CD56+, 80.44%±2.53% were CD8+. The quality requirements for batch release were met in 17 productions. As of the data cut-off date (July 19, 2019), 4 pediatric and 7 adult patients were infused with a single dose of CARCIK-CD19 (n=2 HLA identical sibling, n=4 MUD, n=5 haploidentical donor). The leukemic burden in the BM post lymphodepletion/pre-infusion ranged from 0% to 96%. CARCIK-CD19 were characterized by a high profile of safety in all treated patients. Toxicities reported were a grade I cytokine release syndrome and an infusion-related DMSO-associated seizure, with absence of dose-limiting toxicities, neurotoxicity and graft-versus-host disease (GvHD) in any of the treated patients. Four out of 5 patients, receiving the highest doses, achieved CR and CRi at day 28. The 3 patients in CR were also MRD- (by flow cytometry and RT-PCR) while the CRi was MRD+ and relapsed at day+49. Robust expansion was achieved in the majority of the patients as defined by detectable CAR T-cell detection (vector copy number VCN, range 4645-977992 transgene copies/ug) and flow (range 0.5-30%) in PB. The median time to peak engraftment was 14 days. The magnitude of expansion was correlated with the CD19+ burden in the BM at the time of the infusion (P value = 0.0006, R square 0.7469). CD8+ T cells represented the predominant CARCIK-CD19 T-cell subset (78.88%±5.33% d14 n=6) along with CD3+CD56+ CIK cells and CD4+ T cells to a lesser extent. The majority of CAR T cells had a central and effector memory phenotype. CAR T cells were measurable by VCN up to 6 months with a mean persistence of 70.5 ± 14.85 days (follow up ranging from 28 days to 1 year). No major difference was observed by integration analyses of the patients' PB and the cell products. The vector integration sites reflected the classical random distribution of SB without any tendency for gene dense regions. Conclusions Our ongoing phase I/II trial demonstrates that SB-engineered CARCIK-CD19 cells are able to expand and persist in pediatric and adult B-ALL patients relapsed after HSCT, with important implications for a non-viral technology. These encouraging results prompted us to expand our study. Disclosures Gritti: Autolus Ltd: Honoraria; Roche: Other: Not stated; Abbvie: Other: Not stated; Becton Dickinson: Other: Not stated. Rambaldi:Celgene: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Speakers Bureau; Roche: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Research Funding, Speakers Bureau; Jazz: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau, travel support; Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Gilead: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Speakers Bureau; Amgen: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Research Funding, Speakers Bureau; Novartis: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Speakers Bureau; Italfarmaco: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Research Funding, Speakers Bureau; Omeros: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
- Published
- 2019
- Full Text
- View/download PDF
25. Preclinical Assessment of Non-Virally Engineered CD33.CAR Cytokine-Induced Killer (CIK) Cells in Chemoresistant AML Patient-Derived Xenografts
- Author
-
Chiara F. Magnani, Silvia Arcangeli, Maria Grazia Valsecchi, Ettore Biagi, Marta Serafini, Claudia Cappuzzello, Giuseppe Dastoli, Maria Caterina Rotiroti, Stefania Galimberti, Sarah Tettamanti, Andrea Biondi, Chiara Buracchi, and Zsuzsanna Izsvák
- Subjects
Cytokine-induced killer cell ,business.industry ,medicine.medical_treatment ,Immunology ,CD33 ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Cell therapy ,Leukemia ,medicine.anatomical_structure ,Cytokine ,Antigen ,Aldesleukin ,Cancer research ,medicine ,Bone marrow ,business - Abstract
Background Chimeric Antigen Receptor (CAR)-T cell therapy has been successfully clinically deployed in the context of B-cell malignancies, paving the way for further development also in Acute Myeloid Leukemia (AML), a still unmet clinical need in the field of oncohematology. Among the potential AML targetable antigens, CD33 is so far one of the main validated molecule. Objectives The aim of the present study was to optimize a non-viral gene transfer method to engineer Cytokine-Induced Killer (CIK) cells with a CD33.CAR by using a novel version of the Sleeping Beauty (SB) transposon system, named "SB100X-pT4". Further, a preclinical assessment of SB-modified CD33.CAR-CIK cells was performed in chemoresistant AML Patient-Derived Xenografts (PDX), in order to address the unmet need of targeting drug-resistant AML cells. Methods Donor derived-CIK cells were stably transduced with a CD33.CAR by exploiting the novel hyperactive SB100X transposase and the pT4 transposon (SB100X-pT4). The novel SB system has been in vitro compared to the previous established SB11-pT. In vitro anti-AML activity of CD33.CAR-CIK cells was assessed by flow cytometry-based cytotoxicity (AnnV-7AAD), proliferation (CFSE) and cytokine production (intracellular IFNg and IL2 detection) assays. In vivo efficacy was evaluated in NSG mice transplanted with MA9-NRas AML cell line or PDX samples. A xenograft chemotherapy model mimicking induction therapy ("5+3" Ara-C and doxorubicin) was exploited to examine the potential benefit of CD33.CAR-CIK cells on chemoresistant/residual AML cells. Results By significantly reducing the amount of DNA transposase, the novel SB100X-pT4 combination resulted in higher CAR levels than the SB11-pT. SB100X-pT4-modified CD33.CAR CIK cells showed efficient expansion after 3 weeks (median fold increase of 38.89, n=4). Both transpositions conferred to CD33.CAR-CIK cells a specific killing (up to 70%) against CD33+ AML target cell lines and primary AML cells. The anti-AML proliferative response of SB-modified CD33.CAR-CIK cells was also considerable (up to 70% of CFSE diluted CAR-CIK cells), as well as the cytokine production (up to 35% for IFN-γ and up to 25% for IL-2). To evaluate the effect of SB100X-pT4-modified CD33.CAR-CIK cells particularly on Leukemia Initiating Cells (LICs), CD33.CAR-CIK cells were administered as an "early treatment" in mice transplanted with the MA9-NRas cell line, which retains a high frequency of LICs. At sacrifice, CD33.CAR-CIK cell-treated mice showed a significant bone marrow (BM) engraftment reduction (median 27.80 for the untreated group and 22.60 for the unmanipulated CIK cells vs 6.45 for CD33.CAR-CIK cell, n=4 NSG mice per group, p= 0.02). PDX of two different AML samples at the onset were established to be used as models mimicking different disease conditions. In an "early treatment" model using secondary transplanted PDX, a setting which presumably reflects the typical LIC properties, a clear engraftment reduction in the treated cohort was observed, nearly undetectable in 2/5 mice, as compared to the untreated mice (up to 70% in BM). A significant leukemia reduction was also measured in the peripheral blood and spleen of treated mice, showing CD33.CAR-CIK cell potential of reducing AML dissemination in the periphery. When ex vivo re-exposed to CD33.CAR-CIK cells residual AML cells were still sensitive to the treatment, indicating that no resistance mechanisms occurred. CD33.CAR-CIK cells were also effective in a second model by which the treatment started when AML engraftment was clearly manifested in the BM (> 1%). Finally, when starting CD33.CAR-CIK cell treatment after disease recurrence post induction therapy, a significant disease reduction was observed in the CD33.CAR-CIK-treated group, reaching undetectable levels in half of the mice, as compared to chemotherapy-only treated mice (up to 60% of engraftment in BM)(Figure 1). Conclusions The employment of a non-viral SB-based CD33.CAR-gene transfer approach, which is overall associated to less cumbersome protocols and reduces the cost of goods, offers a unique alternative to current viral-based strategies to be explored in the setting of resistant forms of AML. Disclosures No relevant conflicts of interest to declare.
- Published
- 2019
- Full Text
- View/download PDF
26. Control of murine Ly6Chigh monocyte traffic and immunosuppressive activities by atypical chemokine receptor D6
- Author
-
Massimo Locati, Alberto Mantovani, Achille Anselmo, Nicoletta Caronni, Benedetta Savino, Mauro M. Teixeira, Chiara Buracchi, Adelaida Sarukhan, Federica Benvenuti, Marina G. M. Castor, Raffaella Bonecchi, Vanessa Pinho, Savino, B, Castor, M, Caronni, N, Sarukhan, A, Anselmo, A, Buracchi, C, Benvenuti, F, Pinho, V, Teixeira, M, Mantovani, A, Locati, M, and Bonecchi, R
- Subjects
CCR2 ,Receptors, CCR2 ,Immunology ,Graft vs Host Disease ,Adaptive Immunity ,Biology ,CCL7 ,Biochemistry ,Monocytes ,Mice ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,Immune system ,Immune Tolerance ,medicine ,Animals ,Antigens, Ly ,Scavenger receptor ,CCL13 ,030304 developmental biology ,Inflammation ,Mice, Knockout ,0303 health sciences ,Monocyte ,Cell Biology ,Hematology ,medicine.disease ,3. Good health ,medicine.anatomical_structure ,Graft-versus-host disease ,Gene Expression Regulation ,Receptors, Chemokine ,atypical, chemokine receptors, graft-versus-host disease, immunosuppressive agents, mice, monocytes ,030215 immunology - Abstract
The atypical chemokine receptor D6 is a decoy and scavenger receptor for most inflammatory CC chemokines and prevents the development of exacerbated inflammatory reactions. Here we report that mice lacking D6 expression in the nonhematopoietic compartment have a selective increase in the number of Ly6Chigh monocytes in the circulation and in secondary lymphoid tissues. Under inflammatory conditions, Ly6Chigh monocytes accumulate in increased number in secondary lymphoid organs of D6−/− mice in a CCR2-dependent manner. Ly6Chigh monocytes derived from D6−/− mice have enhanced immunosuppressive activity, inhibit the development of adaptive immune responses, and partially protect mice from the development of GVHD. Thus, control of CCR2 ligands by D6 regulates the traffic of Ly6Chigh monocytes and controls their immunosuppressive potential.
- Published
- 2012
- Full Text
- View/download PDF
27. Targeting a Specific Glycosylated Epitope of CD43 with a New Humanized Monoclonal Antibody for the Treatment of Pediatric and Adult T-Cell Acute Lymphoblastic Leukemia (T-ALL)
- Author
-
Andrea Ghelli Luserna di Rorà, Caterina Riillo, Marco Rossi, Andrea Biondi, Maria Teresa Di Martino, Chiara Buracchi, F. Tuccillo, Maria Cucè, Alessandro Gulino, Claudio Tripodo, Katia Grillone, Maria Anna Siciliano, Pierosandro Tagliaferri, Nicoletta Staropoli, Giuseppe Gaipa, Cirino Botta, Maria Eugenia Gallo Cantafio, Pierfrancesco Tassone, and Giovanni Martinelli
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,biology ,medicine.diagnostic_test ,medicine.drug_class ,business.industry ,Immunology ,Cell Biology ,Hematology ,Monoclonal antibody ,medicine.disease ,Biochemistry ,Epitope ,Flow cytometry ,Leukemia ,Antigen ,Cancer research ,medicine ,biology.protein ,Immunohistochemistry ,Antibody ,business - Abstract
Introduction: T-cell acute lymphoblastic leukemia (T-ALL) accounts for about 20% of pediatric and adult ALL cases. Despite the use of intensive chemotherapy protocols, 25% of children and 50% of adult patients fail to respond or relapse. The 3-years prognosis for these patients is poor and novel treatment options are needed. The targeting of tumor-associated antigens by monoclonal antibodies (mAb) is among the most investigated immune-therapeutic strategies. Accordingly, we developed a new humanized mAb (hUMG1), directed against a heavy glycosylated epitope of CD43 which presents a high reactivity against T-ALL cells. Here we investigated the pre-clinical therapeutic activity and the mechanisms of action of hUMG1 in experimental models of T-ALL. Methods: The expression of hUMG1 target was assessed by flow cytometry on tumor cell lines and primary samples from either T-ALL patients (n=48) or healthy donors (n =6). Humanized mAbs were generated by combining the variable domains of the murine antibody to the corresponding human IgG1 constant domains. Through immunohistochemistry (IHC) we screened several tissue microarrays (TMA) including human, cynomolgusmonkey and macacus rhesus (according to FDA/CE guidelines), rat and mouse normal tissues. Complement-mediated cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) and cellular phagocytosis (ADCP) on T-ALL cells were evaluated by flow cytometry. Six different in vivo models on NSG mice (3 with administration of NK-92-CD16+ effectors) have been generated to evaluate mAbs activity in different disease settings: orthotopic, subcutaneous advanced disease (treatments started at 100 mmc) and subcutaneous limited disease (treatments started the day after injection). Results: By screening different cancer cell lines, we observed hUMG1 target to be highly expressed on malignant T-ALL cells. We then tested T-ALL patient-derived blasts from 48 samples (40 pediatric and 8 adults) collected at diagnosis. The antigen was expressed on 23 out of 48 (48%), and most of them belonged to the subset of cortical (EGIL TIII) T-ALL group. By contrast the target antigen was not expressed on normal bone marrow cells from healthy donors. The analysis of the TMA including human normal tissues revealed a specific binding for thymus cortical lymphocytes only, leading us to hypothesize an acceptable safety profile. A humanized mAb, named hUMG1, and an afucosylated version of this mAb (aUMG1) were then developed. By gene expression profiling, western blot and flow cytometry, we observed that target binding by hUMG does not exert any direct activity on neoplastic cells. Subsequently, to investigate CDC, ADCC or ADCP, T-ALL cells were cultured in the presence of complement, peripheral blood mononuclear cells or macrophages, at increasing concentrations of both antibodies. Neither hUMG1 nor aUMG1 were able to induce CDC on target cells. Conversely, both mAbs induced CD16 downregulation, IFN-g production and degranulation on NK cells (more evident with aUMG1) and significant cytotoxicity against both T-ALL cell lines and primary blasts. Additionally, both mAbs induced ADCP. Lastly we observed a mAb-dependent activation of monocytes in the presence of target cells, as demonstrated by the reduction of CD16+ "non-classical" monocytes. Furthermore, we demonstrated potent activity of both mAbs in different T-ALL in vivo models. In an orthotopic model we observed 5 out of 20 treated mice free of disease after 100 days from injection as compared to none of the control group. In both subcutaneous models, we observed a strong ability of our antibody to delay tumor growth and to increase mice survival. Of note, the addition of NK-92-CD16+ strongly improved the activity of aUMG1. In the attempt to bring this antibody from bench to bedside, we assessed, through IHC, the expression of the hUMG1 target in healthy tissues from cynomolgus monkey, macacus rhesus, rat and mouse. We did not observe any reactivity, suggesting that the mAb target is very specific for human cells. Conclusion: Here, we demonstrated that hUMG1 mAb recognizes an antigen specifically expressed on the majority of T-ALL, and its binding is able to mediate effective cytotoxicity against leukemia in vitro and in vivo, indicating that this antibody, in particular the aUMG1 version, may represent a novel promising immune-therapeutic tool for the treatment of T-ALL patients. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
28. Clinical-Grade Transduction of Allogeneic Cytokine Induced Killer (CIK) Cells with CD19 Chimeric Antigen Receptor (CAR) Using Sleeping Beauty (SB) Transposon: Successful GMP-Compliant Manufacturing for Clinical Applications
- Author
-
Grazia Fazio, Giuseppe Gaipa, Gian Maria Borleri, Valentina Colombo, Attilio Rovelli, Giuseppe Gritti, Silvia Rigamonti, Federico Lussana, Andrea Biondi, Adriana Balduzzi, Giada Matera, Benedetta Cabiati, Giovanni Cazzaniga, Sarah Tettamanti, Stefania Cesana, Eugenio Montini, Chiara Buracchi, Martino Introna, Daniela Belotti, Giuseppe Dastoli, Sara Napolitano, Alessandro Rambaldi, and Chiara F. Magnani
- Subjects
Adoptive cell transfer ,biology ,Cytokine-induced killer cell ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,CD19 ,Chimeric antigen receptor ,Transplantation ,Transduction (genetics) ,Cytokine ,biology.protein ,Cancer research ,Medicine ,business ,Transposase - Abstract
Background: Acute lymphoblastic leukemia (ALL) is a malignant disorder with a long-term remission of less than 50% of adult patients and of nearly 80% of children. Relapsed and refractory (r/r) adult and childhood B-ALL patients, have significant unmet medical needs. Adoptive transfer of patient-derived T cells engineered to express a chimeric antigen receptor (CAR) by viral vectors has achieved complete remission and durable response in highly refractory populations (June CH et al. Science 2018). In addition, unmodified Cytokine Induced Killer (CIK) cells (CD3+, CD56+ T cells) have clearly demonstrated a high profile of safety in ALL patients (Introna M et al. Biol Blood Marrow Transplant. 2017). Here, we demonstrate the feasibility and reproducibility of a GMP-compliant clinical-grade culture and gene-modification protocol of allogeneic CIK cells using the non-viral Sleeping Beauty (SB) transposon system (Singh H et al, Plos One 2013) to obtain CD19CAR expressing CIK cells (Magnani CF et al, Oncotarget 2016, Magnani CF et al, Hum Gene Ther. 2018, Biondi A et al. J Autoimmun. 2017) starting from a limited amount of an easily available material such as peripheral blood (PB). Methods: Fifty mL of PB were centrifuged on Ficoll gradient to obtain mononuclear cells (PBMCs). PBMCs were then simultaneously electro-transferred with the SB GMP-grade DNA transfer CD19.CAR/pTMNDU3 plasmid (human 3rd generation anti-CD19CD28OX40z CAR under the pTMNDU3 promoter), and transposase pCMV-SB11 plasmid (kindly provided by L. Cooper, MDACC, Houston, TX, USA). CIK populations (Introna M et al, Haematologica 2007) were then generated according to the method enclosed in the filed patent EP20140192371 (Magnani CF et al, Oncotarget 2016). The manufacturing process and the quality control tests were performed in a good manufacturing practices (GMP) academic cell factory authorized by Agenzia Italiana del Farmaco (AIFA) in the context of an ongoing phase I clinical trial (NCT03389035) for children and adults with relapsed/refractory B-cell precursor ALL post hematopoietic stem cell transplantation (HSCT). Results: We manufactured nine batches by seeding a mean of 102.52x106 allogeneicPBMCs derived from 50 ml of peripheral blood (range 46.1 - 193.17x106). After 21-22 days of culture the mean harvesting was 5.0x109 nucleated cells (range 1.15 - 16.00x109) with a mean viability of 97.56% (min. 95.24% - max 99.43%). These cells were mostly CD3+ lymphocytes (mean 98.54%, min. 94.85% - max 99.68%) which had a median fold increase of 87.3. Expanded CD3+ cells expressed CD56+ and surface CAR at variable levels among the batches (mean 44.79% and 43.78%, respectively) and had a median vector copy number (VCN) of 2.56 VCN/cells, according to pre-clinical data (Magnani CF et al, Hum Gene Ther. 2018). In all the nine batches, CARCIK-CD19 cells demonstrated potent and specific in vitro cytotoxicity towards the CD19+ REH target cell line (mean 82.96%, min. 61.89% - max 97.72%). Cell products appear to be highly polyclonal and no signs of genotoxicity by transposon insertions could be observed by integration site (IS) analysis performed by Sonication Linker Mediated (SLiM)-PCR and Illumina MiSeq sequencing. The GMP batches were released after about 10 days after the end of production. Quality control release specifications and results are reported in Table 1. Conclusions: Overall, these results demonstrate that clinical-grade SB transduction of allogeneic CIK cells with CD19 CAR is feasible and allows rapid and efficient expansion of highly potent CARCIK-CD19 cells starting from easily available small amounts of PB, with important implications for non-viral technology. In summary our data represent a solid ground for the future development of further SB-based platforms. A clinical trial investigating allogeneic CARCIK-CD19 in r/r pediatric and adult ALL post HSCT is currently ongoing (NCT03389035). Disclosures Gritti: Autolus: Consultancy. Rambaldi:Celgene: Consultancy; Omeros: Consultancy; Novartis: Consultancy; Italfarmaco: Consultancy; Pfizer: Consultancy; Amgen Inc.: Consultancy; Roche: Consultancy.
- Published
- 2018
- Full Text
- View/download PDF
29. Specific Targeting of Acute Myeloid Leukemia By the Use of Non-Virally Engineered CIK (Cytokine-Induced Killer) Cells Expressing the Anti-CD33 Chimeric Antigen Receptor (CAR)
- Author
-
Ettore Biagi, Silvia Arcangeli, Chiara F. Magnani, Maria Caterina Rotiroti, Sarah Tettamanti, Chiara Buracchi, Claudia Cappuzzello, and Andrea Biondi
- Subjects
Cytokine-induced killer cell ,business.industry ,medicine.medical_treatment ,Immunology ,CD33 ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Biochemistry ,03 medical and health sciences ,Haematopoiesis ,Leukemia ,0302 clinical medicine ,medicine.anatomical_structure ,Antigen ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Cytotoxic T cell ,Bone marrow ,business ,030215 immunology - Abstract
Background Acute Myeloid Leukemia (AML) is still associated with high relapse rates when treated with conventional chemotherapeutic and hematopoietic transplantation regimens. Thus, new treatment options are urgently needed. Immunotherapy adopting T cells engineered to express tumor-directed Chimeric Antigen Receptors (CARs) has shown striking results particularly in the context of B-cell malignancies, sparking a keen interest in extending this approach also to other hematological malignancies such as AML. Among the surface molecules identified, the CD33 molecule represents so far one of the main validated target in AML and, being broadly expressed on AML blasts, represents a suitable antigen to be targeted with CAR-T cells. Objectives The aim of the present study is to preclinically evaluate the efficacy and safety profiles of CD33.CAR redirected Cytokine Induced Killer (CIK) cells alone and in combination with standard chemotherapeutic agents. Methods Donor derived- and autologous-CIK cells were stably or transiently transduced with a third generation anti-CD33.CAR by Sleeping Beauty transposon- or mRNA-mediated engineering. In vitro anti-AML activity has been assessed by means of Flow cytometry-based cytotoxicity (AnnV-7AAD staining), proliferation (Ki67 staining and CFSE dilution) and cytokine production (intracellular IFNg and IL2 detection) assays, upon challenge with AML samples. In vivo efficacy has been evaluated in NSG mice transplanted with MA9-NRas AML cell line or primary AML samples. Moreover, an already established xenograft chemotherapy model has been exploited to examine the potential benefit of combining CD33.CAR-CIK cells with standard AML induction therapy (Ara-C and doxorubicin). Results CD33.CAR stably expressing CIK cells were able to induce a potent anti-leukemic activity in vitro, in terms of specific killing either in short term (>70% at 4h, E:T ratio 5:1) and long term cytotoxic assays (>90% at 1 week, E:T ratio 1:10), with statistically significant differences as compared to the unmanipulated condition. Moreover, CD33.CAR-CIK cells were able to retain a significant cytotoxic activity when re-challenged with the CD33+ target following a previous stimulation (up to 65%). The proliferative response to AML target cells was also considerable and CAR-specific (up to 60% of Ki67+CAR-CIK cells and up to 70% of CFSE diluted CAR-CIK cells), as well as the cytokine production (up to 35% of IFN-γ producing CAR-CIK cells and up to 25% of IL-2 producing CAR-CIK cells). CIK cells transiently expressing the CD33.CAR were also effective towards the AML target. In vivo results showed that CD33.CAR-CIK cells were able to control the disease in MA9 grafted mice in all the districts analyzed (peripheral blood, bone marrow, spleen, liver and kidney), as compared to untreated mice. To evaluate the effect of CD33.CAR-CIK cell immunotherapy particularly on Leukemia Initiating Cells (LICs), CD33.CAR-CIK cells were administered as an early treatment approach, treating mice 5 days after i.v. injection of a secondary transplanted PDX sample. We observed a clear engraftment reduction in the treated cohort, nearly undetectable in 2 out 5 mice, while a high leukemic burden has been detected in untreated mice (up to 70% of engraftment in bone marrow). Furthermore, by exploiting CD33.CAR-CIK cell treatment in mice experiencing disease recurrence after the "5+3" chemotherapy-induction protocol, preliminary data showed that CD33.CAR-CIK cells were also capable to target chemotherapy resistant/residual AML cells. Conclusions Considering our in vivo preliminary results, we aim to further evaluate CD33.CAR-CIK cell immunotherapy efficacy, particularly against chemotherapy resistant/residual AML cells. Concerning the safety aspect, since the CD33 targeting raises concerns for a potential myelotoxicity, we will assess the potential long-term off-target effects of CD33.CAR-CIK cells (comparing stably with transiently expressed CD33.CARs) on normal hematopoietic stem/myeloid progenitor cells. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
30. Abstract 1783: UMG1, a novel humanized monoclonal antibody against a glysosylated-CD43-related epitope, induces antibody-dependent cellular cytotoxicity (ADCC) on human T-cell acute lymphoblastic leukemia cells
- Author
-
Marco Rossi, Daniele Caracciolo, Chiara Buracchi, Giuseppe Gaipa, Maria Cucè, F. Tuccillo, Maria Anna Siciliano, Pierfrancesco Tassone, Emanuela Altomare, Pierosandro Tagliaferri, Andrea Biondi, Maria Eugenia Gallo Cantafio, Mariamena Arbitrio, Caterina Riillo, Cirino Botta, and Maria Teresa Di Martino
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,business.industry ,medicine.drug_class ,T cell ,Monoclonal antibody ,Peripheral blood mononuclear cell ,Epitope ,medicine.anatomical_structure ,Oncology ,Antigen ,Cell culture ,Cancer research ,Medicine ,business ,Cytotoxicity - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a hematologic malignancy accounting for about 25% of all acute leukemias. Due to the poor therapeutic scenario and severe prognosis of T-ALL, novel drugs are eagerly awaited. The targeting of tumor-associated antigens by monoclonal antibodies (mAb) for induction of immune-mediated cellular cytotoxicity is presently a promising immunotherapeutic strategy. We previously developed a novel mAb direct against a heavy glycosylated oncofetal epitope of CD43 (UN1) with known potential application as therapeutic and diagnostic tool. By screening different cancer cell lines, we observed that UN1 is highly and selectively expressed on malignant T-ALL cells. The expression of UN1 was then evaluated in 38 T-ALL patient-derived blasts and high correlation for a specific subset of patients (about 80%) belonging to the cortical T-ALL group (EGIL T3) was detected. Accordingly, we developed a humanized mAb, (UMG1) and an afucosylated enginereed version (a-UMG1). Therefore, to elucidate the mechanism of action of these mAbs, we investigated complement-mediated cytotoxicity (CDC), ADCC and antibody-dependent cellular phagocytosis (ADCP). To this aim, T-ALL cells (HPB-ALL and CCRF-CEM) have been cultured in the presence of complement, peripheral blood mononuclear cells (PBMCs), NK-92-CD16+ cells or macrophages, using increasing concentrations of both mAbs. Notably, we observed that mAbs treatment alone did not exert either cellular cytotoxicity or CDC on target cells. Conversely, both mAbs induced significant ADCC mediated by PBMCs and NK-92-CD16+ cell line in term of NK degranulation and cytotoxicity and macrophage-mediated ADCP. In vivo results demonstrated a powerful activity of UMG1 in 3 different models of T-ALL in NSG mice in the presence or absence of NK-92-CD16+ cells. Specifically, in two subcutaneous models, we observed a strong ability of both mAbs to delay tumor growth and increase survival of treated mice. As expected, the combinatory treatment with NK-92-CD16+ cell line strongly improved the activity of a-UMG1. Importantly, in the orthotopic disseminated model, which better reproduces the human T-ALL disease, we observed 5 out of 20 treated mice alive and free of disease 100 days after injection. Furthermore, to explore the possibility of combination therapy, the modulation of UN1 expression by chemotherapeutic agents in T-ALL was investigated. Interestingly, we observed that methotrexate and doxorubicin , alone or in combination, increased UN1 expression at low nanomolar concentrations, and improved ADCC in vitro. In conclusion, we demonstrated that UMG1 and a-UMG1 represent a novel promising immune-therapeutic tool for the treatment of T-ALL patients. Citation Format: Cirino Botta, Maria E. Gallo Cantafio, Chiara Buracchi, Maria A. Siciliano, Maria Cucè, Caterina Riillo, Franca M. Tuccillo, Daniele Caracciolo, Emanuela Altomare, Mariamena Arbitrio, Maria T. Di Martino, Marco Rossi, Andrea Biondi, Giuseppe Gaipa, Pierosandro Tagliaferri, Pierfrancesco Tassone. UMG1, a novel humanized monoclonal antibody against a glysosylated-CD43-related epitope, induces antibody-dependent cellular cytotoxicity (ADCC) on human T-cell acute lymphoblastic leukemia cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1783.
- Published
- 2018
- Full Text
- View/download PDF
31. Differential Effects of Immunosuppressive Drugs on Chemokine Receptor CCR7 in Human Monocyte-Derived Dendritic Cells: Selective Upregulation by Rapamycin
- Author
-
Paola Allavena, Giancarlo Bianchi, Lorenzo Piemonti, Walter Luini, Alessia Mercalli, Giovanna D'Amico, Federica Marchesi, Annunciata Vecchi, Alberto Mantovani, Chiara Buracchi, Cui Hong Yang, Valeria Sordi, Sordi, V, Bianchi, G, Buracchi, C, Mercalli, A, Marchesi, F, D'Amico, G, Yang, Ch, Luini, W, Vecchi, A, Mantovani, A, Allavena, P, Piemonti, Lorenzo, Yang C.,, and Piemonti, L
- Subjects
Receptors, CCR7 ,C-C chemokine receptor type 7 ,Biology ,Monocytes ,Mice ,Chemokine receptor ,Cell Movement ,In vivo ,medicine ,Animals ,Humans ,Antigen-presenting cell ,Migration ,Sirolimus ,Transplantation ,Monocyte ,CCL19 ,Dendritic Cells ,Dendritic cell ,Chemotaxis, Leukocyte ,medicine.anatomical_structure ,Gene Expression Regulation ,Models, Animal ,Immunology ,Cancer research ,Receptors, Chemokine ,Lymph Nodes ,Immunosuppressive Agents ,Immunosuppression ,Ex vivo - Abstract
BACKGROUND. Appropriate recruitment of dendritic cells (DC) at sites of inflammation and migration to secondary lymphoid organs is of critical importance for the initiation of Ag-specific immune responses. The proper localization of DC in selected tissues is guided primarily by the coordinated expression of chemokine receptors (CKR). Here we show that immunosuppressive drugs have divergent effects on the modulation of CKR in maturing DC. METHODS AND RESULTS. Dexamethazone (DEX) and IL-10 inhibited human DC migration to CCL19 in vitro and mouse DC migration to lymph nodes (LN) in vivo, by impairing CCR7 expression. The calcineurin inhibitors cyclosporine A (CsA) and tacrolimus (FK506) were characterized by the inability to modulate CKR expression and migratory activity. Rapamycin (RAPA) increased DC migration to CCL19 in vitro and to LN in vivo by enhancing CCR7 expression. This effect could be mediated, in LPS-maturing DC, by the inhibition of autocrine IL-10 production. The in vivo data obtained with ex vivo RAPA treated DC were confirmed in a model of in vivo drug administration in mice, suggesting a potential clinical relevance. CONCLUSIONS. These findings demonstrate that immunosuppressive agents differently modulate the CKR switch associated with maturing DC; in particular, RAPA selectively up-regulates CCR7 and enhances the migration of differentiated DC to regional LN. This study contributes to a better understanding of the role of immunosuppressive therapy on DC migration, a potentially relevant check point of immunosuppressive treatment.
- Published
- 2006
- Full Text
- View/download PDF
32. Increased inflammation in mice deficient for the chemokine decoy receptor D6
- Author
-
Luca Vago, Raffaella Bonecchi, Daniel Rukavina, Jana Dupor, Sergio A. Lira, Massimo Locati, Yeny Martinez de la Torre, Annunciata Vecchi, Alberto Mantovani, Donald N. Cook, Chiara Buracchi, and Manuela Nebuloni
- Subjects
Inflammation ,CCR1 ,Chemokine ,biology ,Immunology ,Mice, Transgenic ,Receptors, CCR10 ,Cell biology ,Mice ,Chemokine receptor ,biology.protein ,medicine ,Animals ,Immunology and Allergy ,Receptors, Chemokine ,Lymph Nodes ,Decoy receptors ,medicine.symptom ,Scavenger receptor ,Decoy ,Receptor ,Skin - Abstract
Chemokines are chemotactic cytokines with a key role in the control of cell trafficking and positioning under homeostatic and inflammatory conditions. D6 is a promiscuous 7-transmembrane-domain receptor expressed on lymphatic vessels which recognizes most inflammatory, but not homeostatic, CC chemokines. In vitro experiments demonstrated that D6 is unable to signal after ligand engagement, and it is structurally adapted to sustain rapid and efficient ligand internalization and degradation. These unique functional properties lead to the hypothesis that D6 may be involved in the control of inflammation by acting as a decoy and scavenger receptor for inflammatory chemokines. Consistent with this hypothesis, here we report that D6(-/-) mice showed an anticipated and exacerbated inflammatory response in a model of skin inflammation. Moreover, the absence of D6 resulted in increase cellularity and inflammatory-chemokine levels in draining lymph nodes. Thus, D6 is a decoy receptor structurally adapted and strategically located to tune tissue inflammation and control transfer of inflammatory chemokines to draining lymph nodes.
- Published
- 2005
- Full Text
- View/download PDF
33. Megakaryocytes contribute to the bone marrow-matrix environment by expressing fibronectin, type IV collagen, and laminin
- Author
-
Gianluca Viarengo, David L. Kaplan, Manuela Currao, Chiara Buracchi, Cristian Gruppi, Alessandra Balduini, Giuseppe Celesti, Luigi Laghi, Alessandro Malara, Malara, A, Currao, M, Gruppi, C, Celesti, G, Viarengo, G, Buracchi, C, Laghi, L, Kaplan, D, and Balduini, A
- Subjects
Collagen Type IV ,Myelosuppression ,Antimetabolites ,Article ,Extracellular matrix ,Type IV collagen ,Mice ,Megakaryocyte ,Laminin ,Bone Marrow ,medicine ,Animals ,Stem Cell Niche ,Thrombopoietin ,biology ,Cell Biology ,Cell biology ,Fibronectins ,Fibronectin ,Haematopoiesis ,medicine.anatomical_structure ,Gene Expression Regulation ,Immunology ,biology.protein ,Molecular Medicine ,Bone marrow ,Fluorouracil ,Megakaryocytes ,Developmental Biology - Abstract
Megakaryocytes associate with the bone marrow vasculature where they convert their cytoplasm into proplatelets that protrude through the vascular endothelium into the lumen and release platelets. The extracellular matrix (ECM) microenvironment plays a critical role in regulating these processes. In this work we demonstrate that, among bone marrow ECM components, fibronectin, type IV collagen, and laminin are the most abundant around bone marrow sinusoids and constitute a pericellular matrix surrounding megakaryocytes. Most importantly, we report, for the first time, that megakaryocytes express components of the basement membrane and that these molecules contribute to the regulation of megakaryocyte development and bone marrow ECM homeostasis both in vitro and in vivo. In vitro, fibronectin induced a threefold increase in the proliferation rate of mouse hematopoietic stem cells leading to higher megakaryocyte output with respect to cells treated only with thrombopoietin or other matrices. However, megakaryocyte ploidy level in fibronectin-treated cultures was significantly reduced. Stimulation with type IV collagen resulted in a 1.4-fold increase in megakaryocyte output, while all tested matrices supported proplatelet formation to a similar extent in megakaryocytes derived from fetal liver progenitor cells. In vivo, megakaryocyte expression of fibronectin and basement membrane components was upregulated during bone marrow reconstitution upon 5-fluorouracil induced myelosuppression, while only type IV collagen resulted upregulated upon induced thrombocytopenia. In conclusion, this work demonstrates that ECM components impact megakaryocyte behavior differently during their differentiation and highlights a new role for megakaryocyte as ECM-producing cells for the establishment of cell niches during bone marrow regeneration. Stem Cells 2014;32:926–937
- Published
- 2013
34. Euroflow-Based Immunophenotypic Characterization of CD34+ Cell Compartment in Juvenile Myelomonocytic Leukemia (JMML): A New Tool for Differential Diagnosis
- Author
-
Sergio Matarraz, Elaine Sobral da Costa, Alberto Orfao, Ester Mejstrikova, Andrea Biondi, Vincent H.J. van der Velden, Carmen Mariana Aanei, Giuseppe Gaipa, Lukasz Sedek, Chiara Buracchi, Alita van der Sluijs, Tomasz Szczepański, Jacques J.M. van Dongen, Cristina Bugarin, and Michaela Novakova
- Subjects
Pathology ,medicine.medical_specialty ,Myeloid ,Juvenile myelomonocytic leukemia ,biology ,CD117 ,business.industry ,Immunology ,CD33 ,CD34 ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Leukemia ,medicine.anatomical_structure ,Immunophenotyping ,medicine ,biology.protein ,Bone marrow ,business - Abstract
Background Juvenile myelomonocytic leukemia (JMML) is a lethal myeloproliferative disease (MPD) of young childhood characterized by an overproduction of myelomonocytic cells and an increased in vitro sensitivity of hematopoietic progenitors to granulocyte-macrophage colony-stimulating factor [GM-CSF] (Emanuel PD et al, Blood 1991). Diagnostic criteria for JMML are currently well-established and based on clinical features and laboratory findings. However, in some patients diagnosis of JMML vs other overlapping disease entities, still remains a challenge, immunophenotyping being not part of the diagnostic work-up of JMML. Here, we aimed at detailed characterization of the CD34+ cell compartment in JMML bone marrow (BM) using the standardized EuroFlow myeloid panel in combination with innovative EuroFlow software maturation tools. Our major goal was to determine the potential utility of immunophenotyping of CD34 cells in the diagnostic work-up of JMML. Methods Overall, we analyzed BM cells from 10 JMML patients at diagnosis (age range: 0-7 years), 17 control subjects (age range: 0-15 years) and 5 patients (age range: 0-5 years) with a suspected diagnosis of JMML that was subsequently not confirmed following standardized EuroFlow antibody combinations: 1) cyCD3/ CD45/ cyMPO/ cyCD79a/ CD34/ CD19 / CD7/smCD3 (for early lineage assignement); 2) HLADR/CD45/CD16/CD13/CD34/CD117/CD11b/CD10 (neutrophilic maturation); 3) HLADR/CD45/CD35/CD64/CD34/CD117/CD300e (IREM2)/CD14 (monocytic maturation); 4) HLADR/CD45/CD36/CD105/CD34/CD117/CD33/CD71 (erythroid vs plasmacytoid dendritic cell maturation). Samples were processed and analyzed according to the Euroflow standard operating protocols (van Dongen JJM et al, Leukemia 2012, Kalina T et al, Leukemia 2012). Data analysis was specifically focused on the immunophenotypic profile of CD34+ gated cells. Results Within the CD34+ BM cell compartment the proportion (mean % ± 1SD) of granulocytic and monocytic precursors were not significantly different in JMML as compared to controls: 33% ± 15% vs 25% ± 12% (p = 0.16) and 14% ± 6.3% vs 12% ± 7.1% (p = 0.68) respectively. Otherwise we observed a slightly decreased in erythroid CD34+ progenitors in JMML vs controls (1.0% ± 1.2% vs 2.8% ± 1.7%, p Conclusions CD34+ precursor cells from JMML patients display a unique immunophenotypic profile characterized by an inverted ratio of CD19+ B/CD7+ lymphoid precursors, associated with unusual marker coexpressions, which might contribute to fast and more precise diagnostic work-up of JMML. Further studies in larger patient series are required to confirm our observations. Disclosures Biondi: Novartis: Membership on an entity's Board of Directors or advisory committees, Other: Advisory Board; BMS: Membership on an entity's Board of Directors or advisory committees; Cellgene: Other: Advisory Board.
- Published
- 2016
- Full Text
- View/download PDF
35. Role of the chemokine scavenger receptor D6 in balancing inflammation and immune activation
- Author
-
Elena M, Borroni, Chiara, Buracchi, Benedetta, Savino, Fabio, Pasqualini, Remo C, Russo, Manuela, Nebuloni, Raffaella, Bonecchi, Alberto, Mantovani, and Massimo, Locati
- Subjects
Inflammation ,Mice, Knockout ,Microscopy, Confocal ,CHO Cells ,Mycobacterium tuberculosis ,Receptors, CCR10 ,Immunohistochemistry ,Mice ,Cricetulus ,Cricetinae ,Immune System ,Animals ,Humans ,Tuberculosis - Abstract
Chemokines play a major role in the induction of inflammatory reactions and development of an appropriate immune response by coordinating leukocyte recruitment. The appropriate control of the chemokine system involves several chemokine decoy receptors, with distinct specificity and tissue distribution, defined as nonactivating chemokine receptors able to bind the ligands and target them to degradation. The best-characterized representative of these receptors is D6, which is located on lymphatic endothelium and controls most inflammatory CC chemokines. Here we will discuss the expression and regulation of D6 during challenge with the pathogen, and its role in dampening inflammation in tissues and draining lymph nodes and in the organization of a protective immune response.
- Published
- 2009
36. Chapter 11 Role of the Chemokine Scavenger Receptor D6 in Balancing Inflammation and Immune Activation
- Author
-
Alberto Mantovani, Chiara Buracchi, Massimo Locati, Benedetta Savino, Raffaella Bonecchi, Manuela Nebuloni, Elena Monica Borroni, Remo C. Russo, and Fabio Pasqualini
- Subjects
CCR1 ,Chemokine receptor ,Immunology ,CCR10 ,CXC chemokine receptors ,C-C chemokine receptor type 6 ,Biology ,CCL13 ,CC chemokine receptors ,CCL21 - Abstract
Chemokines play a major role in the induction of inflammatory reactions and development of an appropriate immune response by coordinating leukocyte recruitment. The appropriate control of the chemokine system involves several chemokine decoy receptors, with distinct specificity and tissue distribution, defined as nonactivating chemokine receptors able to bind the ligands and target them to degradation. The best‐characterized representative of these receptors is D6, which is located on lymphatic endothelium and controls most inflammatory CC chemokines. Here we will discuss the expression and regulation of D6 during challenge with the pathogen, and its role in dampening inflammation in tissues and draining lymph nodes and in the organization of a protective immune response.
- Published
- 2009
- Full Text
- View/download PDF
37. TNF-alpha and the IFN-gamma-inducible protein 10 (IP-10/CXCL-10) delivered by parvoviral vectors act in synergy to induce antitumor effects in mouse glioblastoma
- Author
-
Jean Rommelaere, Silvano Sozzani, S Paschek, Jj Cornelis, J. Van Damme, Sofie Struyf, Fabian Kiessling, Annunciata Vecchi, Chiara Buracchi, M Enderlin, R Kinscherf, Ev Kleinmann, and Christiane Dinsart
- Subjects
H-1 parvovirus ,Cancer Research ,medicine.medical_treatment ,Genetic Vectors ,Mice ,Necrosis ,Immune system ,In vivo ,Glioma ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,Vector (molecular biology) ,Molecular Biology ,biology ,Tumor Necrosis Factor-alpha ,Drug Synergism ,Dendritic Cells ,biology.organism_classification ,medicine.disease ,In vitro ,Chemokine CXCL10 ,Mice, Inbred C57BL ,Cytokine ,Immunology ,Cancer research ,Minute Virus of Mice ,Molecular Medicine ,Tumor necrosis factor alpha ,Female ,Glioblastoma ,Immunocompetence ,Minute virus of mice - Abstract
Interferon-gamma-inducible protein 10 is a potent chemoattractant for natural killer cells and activated T lymphocytes. It also displays angiostatic properties and some antitumor activity. Tumor necrosis factor-alpha (TNF-alpha) is a powerful immunomodulating cytokine with demonstrated tumoricidal activity in various tumor models and the ability to induce strong immune responses. This prompted us to evaluate the antitumor effects of recombinant parvoviruses designed to deliver IP-10 or TNF-alpha into a glioblastoma. When Gl261 murine glioma cells were infected in vitro with an IP-10- or TNF-alpha-transducing parvoviral vector and were subcutaneously implanted in mice, tumor growth was significantly delayed. Complete tumor regression was observed when the glioma cells were coinfected with both the vectors, demonstrating synergistic antitumor activity. In an established in vivo glioma model, however, repeated simultaneous peritumoral injection of the IP-10- and TNF-alpha-delivering parvoviruses failed to improve the therapeutic effect as compared with the use of a single cytokine-delivering vector. In this tumor model, cytokine-mediated immunostimulation, rather than inhibition of vascularization, is likely responsible for the therapeutic efficacy.
- Published
- 2008
38. Non-signaling chemokine receptors: mechanism of action and role in vivo
- Author
-
Massimo Locati, Raffaella Bonecchi, Alberto Mantovani, Chiara Buracchi, Elena Monica Borroni, and Benedetta Savino
- Subjects
CCR1 ,Immunology ,Immunity ,Models, Immunological ,Biology ,Cell biology ,Chemokine receptor ,CXCL2 ,Neurology ,Cell Movement ,Immunology and Allergy ,XCL2 ,Animals ,Humans ,Receptors, Chemokine ,Neurology (clinical) ,CXC chemokine receptors ,CCL25 ,CCL13 ,CCL21 - Abstract
Cell migration is fundamental for numerous biological processes and is critical for the pathogenesis of several diseases. Chemokines represent the main class of mediators providing cell directional migration and several levels of regulation of their function have been identified. A subfamily of chemokine receptors not able to transduce chemotactic signals plays an important role in the control of chemokine concentrations through binding, internalization and degradation of chemotactic factors. Here we review in vitro and in vivo evidences indicating that these 'silent' chemokine receptors represent a strategy to regulate innate and adaptive immunity.
- Published
- 2008
39. Protection against inflammation- and autoantibody-caused fetal loss by the chemokine decoy receptor D6
- Author
-
Yeny Martinez de la Torre, Manuela Nebuloni, Roberta Bulla, Andrea Doni, Daniel Rukavina, Sergio A. Lira, Jana Dupor, Fabio Pasqualini, Pier Luigi Meroni, Eleonora Lauri, Massimo Locati, Bodduluri Haribabu, Luca Vago, Annunciata Vecchi, Alberto Mantovani, Chiara Buracchi, Chiara Agostinis, Elena Monica Borroni, Donald N. Cook, Francesco Tedesco, and Raffaella Bonecchi
- Subjects
Lipopolysaccharides ,Chemokine ,Placenta ,Inflammation ,Receptors, CCR10 ,trophoblast ,leukocyte ,placenta ,decoy receptor D6 ,Settore MED/08 - Anatomia Patologica ,Mice ,Chemokine receptor ,Syncytiotrophoblast ,Pregnancy ,Leukocytes ,medicine ,Animals ,CCR10 ,Fetal Death ,reproductive and urinary physiology ,Mice, Knockout ,Settore MED/04 - Patologia Generale ,Multidisciplinary ,Keywords:trophoblast ,biology ,Autoantibody ,Trophoblast ,Biological Sciences ,Trophoblasts ,Settore MED/16 - Reumatologia ,medicine.anatomical_structure ,Chemokines, CC ,embryonic structures ,Immunology ,Antibodies, Antiphospholipid ,biology.protein ,Female ,Receptors, Chemokine ,medicine.symptom - Abstract
Fetal loss in animals and humans is frequently associated with inflammatory conditions. D6 is a promiscuous chemokine receptor with decoy function, expressed in lymphatic endothelium, that recognizes and targets to degradation most inflammatory CC chemokines. Here, we report that D6 is expressed in placenta on invading extravillous trophoblasts and on the apical side of syncytiotrophoblast cells, at the very interface between maternal blood and fetus. Exposure of D6 −/− pregnant mice to LPS or antiphospholipid autoantibodies results in higher levels of inflammatory CC chemokines and increased leukocyte infiltrate in placenta, causing an increased rate of fetal loss, which is prevented by blocking inflammatory chemokines. Thus, the promiscuous decoy receptor for inflammatory CC chemokines D6 plays a nonredundant role in the protection against fetal loss caused by systemic inflammation and antiphospholipid antibodies.
- Published
- 2007
40. Damping excessive inflammation and tissue damage in Mycobacterium tuberculosis infection by Toll IL-1 receptor 8/single Ig IL-1-related receptor, a negative regulator of IL-1/TLR signaling
- Author
-
Nadia Caccamo, Francesco Dieli, Cecilia Garlanda, Marco Pio La Manna, Diana Di Liberto, Alfredo Salerno, Annunciata Vecchi, Alberto Mantovani, Chiara Buracchi, GARLANDA C, DI LIBERTO D, VECCHI A, LA MANNA MP, BURACCHI C, CACCAMO N, SALERNO A, DIELI F, and MANTOVANI A
- Subjects
Tuberculosis ,Neutrophils ,Immunology ,Interleukin-1beta ,Inflammation ,Biology ,Peripheral blood mononuclear cell ,Antibodies ,Mycobacterium tuberculosis ,Mice ,Necrosis ,Cell Movement ,Macrophages, Alveolar ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Receptor ,Lung ,Tuberculosis, Pulmonary ,Tumor Necrosis Factor-alpha ,Toll-Like Receptors ,Receptors, Interleukin-1 ,Dendritic Cells ,medicine.disease ,biology.organism_classification ,In vitro ,Mice, Mutant Strains ,medicine.anatomical_structure ,Liver ,Cytokines ,medicine.symptom ,Toll IL-1 Receptor 8/Single Ig IL-1-Related Receptor, Inlfammation, Mycobacterium tuberculosis ,Interleukin-1 ,Signal Transduction - Abstract
Toll IL-1R 8/single Ig IL-1-related receptor (TIR8/SIGIRR) is a member of the IL-1R family, expressed by epithelial tissues and immature dendritic cells, and is regarded as a negative regulator of TLR/IL-1R signaling. Tir8-deficient mice were rapidly killed by intranasal administration of low doses of Mycobacterium tuberculosis, despite controlling efficiently the number of viable bacilli in different organs. Tir8−/−-infected mice showed an increased number of neutrophils and macrophages in the lungs; however, mycobacteria-specific CD4 and CD8 T cells were similar in Tir8−/− and Tir8+/+ mice. Exaggerated mortality of Tir8−/− mice was due to massive liver necrosis and was accompanied by increased levels of IL-1β and TNF-α in lung mononuclear cells and serum, as well as by increased production of IL-1β and TNF-α by M. tuberculosis-infected dendritic cells in vitro. Accordingly, blocking IL-1β and TNF-α with a mix of anti-cytokine Abs, significantly prolonged survival of Tir8−/− mice. Thus, TIR8/SIGIRR plays a key role in damping inflammation and tissue damage in M. tuberculosis infection.
- Published
- 2007
41. Mass Cytometry Analysis Dissects CRLF2-Driven Signaling Pathways in Childhood B-Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL)
- Author
-
Kara L. Davis, Daniela Silvestri, Giuseppe Gaipa, Andrea Biondi, Angela Maria Savino, Stefania Pinto, Garry P. Nolan, Chiara Buracchi, Jolanda Sarno, Cristina Bugarin, Martin J. S. Dyer, Chiara Palmi, Ruth C. Barber, Astraea Jager, and Gianni Cazzaniga
- Subjects
MAPK/ERK pathway ,education.field_of_study ,Immunology ,Population ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Dasatinib ,Leukemia ,medicine.anatomical_structure ,medicine ,Cancer research ,biology.protein ,education ,Tyrosine kinase ,PI3K/AKT/mTOR pathway ,B cell ,STAT5 ,medicine.drug - Abstract
BACKGROUND: Rearrangements of the CRLF2 gene, present in 7-15% of childhood BCP-ALL, are responsible of the overexpression of Thymic Stromal Lymphopoietin Receptor (TSLPR) and they are correlated with poor prognosis (Chen IM Blood 2012). TSLPR overexpression can be associated with JAK2 mutations, which leads to aberrant activation of JAK/STAT and PI3K/AKT pathways. Although the cross talk of the signaling pathways is still under investigation, there is a rationale for the use of targeted tyrosine kinase inhibitors (TKIs) to treat this subgroup of patients (Maude SL Blood 2012). We focused on the dissection of CRLF2-driven signaling in primary CRLF2 rearranged(r) BCP-ALL samples by using single cell mass cytometry (CyTOF) analysis. We leveraged the high dimensional single cell capability of the CyTOF to understand, with previously unattainable resolution, the activation of these pathways simultaneously in single cells and their response to inhibition with TKIs and anti-TSLPR monoclonal antibodies (mAbs). This revealed heterogeneity in signaling response, identifying subpopulations which differentially activate intracellular signals through TSLPR and differentially respond to ex vivo treatment. METHODS: Twelve BCP-ALL primary samples, 6 CRLF2r and 6 CRLF2 wild type (wt), were investigated and the expression of 24 phenotypic and 15 functional proteins were measured at single cell level using CyTOF as previous described (Bendall SC Science 2011). To assess the response to ex-vivo TSLP stimulation (10ng/mL) and TKIs/mAbs treatment, data were normalized to the basal levels of each phosphoprotein and significance was calculated using student`s t-test. One million cells per condition were treated with different TKIs, Dasatinib, Ruxolitinib and BEZ-235, and two different clones of anti-TSLPR mAbs (130A10 and 130H3) from MRC Technology. RESULTS: As expected, we observed an aberrant TSLP-induced activation of pSTAT5 and prpS6 in CRLF2r patients as compared with CRLF2wt, used as control group (p=0.0055, p= 0.0007). Of note, we also observed a previously not described TSLP-dependent activation of pERK and pCREB (p=0.0313, p=0.0261) suggesting a cross-talk of the TSLPR-driven signaling also with the RAS/MEK pathway. Treatment with TKIs revealed strong inhibitory activity of Dasatinib, which completely inhibited the TSLP-mediated phosphorylation of STAT5, rpS6, CREB and ERK in CRLF2r treated blasts compared to CRLF2r not treated cells (p= 0.0040, 0.0017, 0.0007, 0.0114 respectively). Ruxolitinib, JAK1/2 inhibitor, also reduced rpS6, CREB and ERK phosphorylation (p=0.0025, 0.037, 0.0132). Interestingly one of the two anti-TSLPR tested mAbs (130A10) was also able to significantly inhibit the TSLP-mediated activation of STAT5, rpS6, and ERK (p= 0.0071, 0.0006, 0.0323). Finally, the PI3K/TORk inhibitor, BEZ-235, did not show any statistically significant reductions. Single cell analysis revealed a population of TSLPR overexpressing blasts (range 20-50%) in which the TSLP stimulation resulted in activation of prpS6 but not pSTAT5, present in all the CRLF2r patients. This rpS6 activation could be inhibited by anti-TSLPR mAb, Dasatinib, Ruxolitinib and BEZ-235, except for one patient in which the activation was blunted only by anti-TSLPR mAb and Dasatinib suggesting an activation of prpS6 through a non canonical pathway. This data reveals heterogeneous signaling populations present within this subtype of leukemia driven by TSLPR overexpression. Finally in 3 additional CRLF2r primary samples, we investigated signaling profile of residual blasts (MRD) at Day8 and Day15 post induction initiation. TSLPR expression was consistently maintained in all patients at both time points. Furthermore, residual blasts were still able to respond to TSLP and the induced pSTAT5 could be effectively inhibited by 130A10 anti-TSLPR clone and Ruxolitinib. CONCLUSION: In summary, these data suggest heterogeneity of TSLPR-related signaling with activation of the expected JAK/STAT and PI3K pathways but also RAS/MEK and CREB activation. Further, TSLPR+ blasts exhibit heterogeneous responses to both treatment with TSLP in combination with TKIs or mAb. Finally, the MRD detection by CyTOF allowed the study of the functional activity of the TSLPR positive resistant cells suggesting a role of CRLF2r in the persistence of the leukemic cells and its targeting to treat late and refractory stages of the disease. Disclosures Davis: Fluidigm, Inc: Honoraria. Dyer:Roche Pharmaceuticals: Speakers Bureau; Gilead: Research Funding; ONO Pharmaceuticals: Research Funding. Nolan:Fluidigm, Inc: Equity Ownership.
- Published
- 2015
- Full Text
- View/download PDF
42. The chemoattractant decoy receptor D6 as a negative regulator of inflammatory responses
- Author
-
Massimo Locati, Y. Martinez de la Torre, Annunciata Vecchi, Alberto Mantovani, Chiara Buracchi, Raffaella Bonecchi, Emanuela Galliera, and Elena Monica Borroni
- Subjects
Settore MED/04 - Patologia Generale ,CCR1 ,Inflammation ,CCR2 ,biology ,Chemokine receptor CCR5 ,Receptors, Interleukin-1 ,C-C chemokine receptor type 6 ,Receptors, CCR10 ,Ligands ,Biochemistry ,Models, Biological ,Cell biology ,Chemokine receptor ,Immunology ,biology.protein ,Leukocytes ,Humans ,Receptors, Chemokine ,CCL13 ,CC chemokine receptors ,CCL21 ,Lymphatic Vessels - Abstract
Other than signalling receptors sustaining leucocyte recruitment during inflammatory reactions, the chemokine system includes ‘silent’ receptors with distinct specificity and tissue distribution. The best-characterized molecule of this subgroup is the CC chemokine receptor D6, which binds most inflammatory CC chemokines and targets them to degradation via constitutive ligand-independent internalization. Structure–function analysis and recent results with gene-targeted animals indicate that D6 has unique functional and structural features, which make it ideally adapted to act as a chemokine decoy and scavenger receptor, strategically located on lymphatic endothelium and placenta to dampen inflammation in tissues and draining lymph nodes.
- Published
- 2006
43. Intestinal inflammation in mice deficient in Tir8, an inhibitory member of the IL-1 receptor family
- Author
-
Maida De Bortoli, Emilio Hirsch, Annunciata Vecchi, Alberto Mantovani, Raffaella Bergottini, Marina Sironi, Chiara Buracchi, Cecilia Garlanda, Nadia Polentarutti, Federica Riva, Eugenio Scanziani, Marta Muzio, Garlanda, C, Riva, F, Polentarutti, N, Buracchi, C, Sironi, M, De Bortoli, M, Muzio, M, Bergottini, R, Scanziani, E, Vecchi, A, Hirsch, E, and Mantovani, A
- Subjects
Lipopolysaccharides ,protein Tir8 ,Lipopolysaccharide ,CpG Oligodeoxynucleotide ,Receptors, Cell Surface ,Inflammation ,Biology ,TIR-8 ,Mice ,chemistry.chemical_compound ,Intestinal mucosa ,inflammatory bowel disease ,interleukin 1 receptor ,medicine ,Animals ,RNA, Messenger ,Intestinal Mucosa ,Receptor ,innate immunity ,Mice, Knockout ,Membrane Glycoproteins ,Multidisciplinary ,Innate immune system ,oligodeoxynucleotide ,Toll-Like Receptors ,lipopolysaccharide ,Receptors, Interleukin-1 ,toll like receptor ,Dendritic Cells ,Biological Sciences ,Enteritis ,Cell biology ,unclassified drug ,Mice, Inbred C57BL ,Interleukin 10 ,chemistry ,Cytokines ,Chemokines ,medicine.symptom ,Signal transduction - Abstract
TIR8, also known as single Ig IL-1-related receptor, is a member of the IL-1 receptor/Toll-like receptor (TLR) superfamily, which acts as an intracellular decoy for components of the signaling pathway. Here we report that Tir8 has a unique pattern of expression, which includes mucosal tissues and dendritic cells (DC). Tir8 -deficient DC showed increased cytokine production in response to TLR agonists (lipopolysaccharide, CpG oligodeoxynucleotides). Tir8 -deficient mice had normal susceptibility to systemic lipopolysaccharide toxicity and to i.p. or s.c. inflammation. However, Tir8 -deficient mice were more susceptible to intestinal inflammation. Thus, TIR8 represents a negative pathway of regulation of the IL-1 receptor/TLR system, expressed in epithelial cells and DC, crucial for tuning inflammation in the gastrointestinal tract.
- Published
- 2004
44. Loss of PTEN in Pediatric T-Cell Acute Lymphoblastic Leukemia Patients: Proteomic and Molecular Characterization
- Author
-
Cristina Bugarin, Grazia Fazio, Maddalena Paganin, Luca Lo Nigro, Giuseppe Gaipa, Giovanni Cazzaniga, Andrea Biondi, Giuseppe Basso, Chiara Buracchi, and Paola Bonaccorso
- Subjects
Mutation ,Immunology ,Wild type ,Cell Biology ,Hematology ,Biology ,medicine.disease_cause ,medicine.disease ,Biochemistry ,Exon ,Leukemia ,Cancer research ,medicine ,biology.protein ,Tensin ,PTEN ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
Signaling networks such as the PI3K/Akt/PTEN/mTOR pathway play a role in the modulation of the aggressiveness of T-Cell Acute Lymphoblastic Leukemia (T-ALL), but the significance of an aberrant activation of this pathway is poorly investigated. The master regulator of PI3K-AKT signaling is the lipid phosphatase and tensin homolog (PTEN); decreased or absent PTEN expression or activity could be associated with a constitutive activation of PI3K/Akt/PTEN/mTOR signaling pathway in T-ALLs. We investigated PTEN Exon7 mutations in diagnostic DNA samples from 67 pediatric T-ALL enrolled in AIEOP ALL2000 and R2006 protocols according to methods by Bandapalli et al. (Haematologica, 2012). We also investigated the PI3K/Akt/PTEN/mTOR pathway using Western Blotting (WB) and Flow Cytometry (FC) in parallel. WB analysis was performed using standard RIPA buffer for protein extraction. FC analysis of protein expression was applied as previously described (Gaipa G et al, Leukemia 2008), and protein levels were measured as % of positive cells compared to isotype control. Positivity or negativity by WB was established by presence or absence of a protein band, while for FC a threshold for positivity was set at ≥ 1% of positive cells. PTEN Exon 7 mutations were identified in 11 out of 67 (16.4%) of patients. According to samples availability, PI3K/Akt/PTEN/mTOR pathway was studied in 9 out of 67 patients (3 PTEN Exon 7 mutated and 6 wild type). PTEN protein resulted completely absent in all three PTEN Exon 7 mutated patients, by contrast PTEN was expressed in all 6 PTEN exon 7 wild type patients (mean by FC 46.98% ± 28.58%). This finding was fully confirmed when WB was applied to the same samples. We did not observe any statistically significant differences in p4EBP1 or mTOR levels in Exon 7 mutated patients as compared to wild type. By contrast, PTEN Exon 7 mutated blasts showed lower phospho-S6 levels compared to PTEN Exon7 wild type patients (mean 3.0% of positive cells vs 32.0%, p=ns). In conclusion, our data show a frequency of PTEN Exon 7 mutations of 16.4%, in agreement with the report by Bandapalli et al. (17.3%). Interestingly, we observed a strong association between the PTEN Exon 7 mutation and the total absence of PTEN protein in the pediatric T-ALL patients studied (data not reported so far to our knowledge). Moreover, although the differences are not statistically significant, p-S6 expression resulted consistently lower in mutated patients as compared to wild type patients. If confirmed in a larger cohort of pediatric T-ALLs, our data could open new insights in the significance of PTEN Exon 7 mutation in pediatric T-ALL and its associated functional profile. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
45. Functional TRAIL receptors in monocytes and tumor-associated macrophages: A possible targeting pathway in the tumor microenvironment
- Author
-
Manuela Liguori, Cristina Belgiovine, Marina Sironi, Paola Allavena, Francesca Bergomas, Fabio Grizzi, Samantha Pesce, Fabio Pasqualini, Alberto Mantovani, Chiara Buracchi, Liguori, M, Buracchi, C, Pasqualini, F, Bergomas, F, Pesce, S, Sironi, M, Grizzi, F, Mantovani, A, Belgiovine, C, and Allavena, P
- Subjects
0301 basic medicine ,TRAIL ,Inflammation ,Monocytes ,law.invention ,TNF-Related Apoptosis-Inducing Ligand ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,law ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,TRAIL receptors ,Molecular Targeted Therapy ,Fibrosarcoma ,Receptor ,Cells, Cultured ,Tumor microenvironment ,tumor-associated macrophages ,business.industry ,Macrophages ,apoptosis ,TRAIL , tumor-associated macrophages ,medicine.disease ,In vitro ,Mice, Inbred C57BL ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,030104 developmental biology ,Oncology ,chemistry ,Apoptosis ,Apigenin ,Immunology ,Recombinant DNA ,targeting macrophages ,medicine.symptom ,business ,Signal Transduction ,Research Paper - Abstract
// Manuela Liguori 1 , Chiara Buracchi 1 , Fabio Pasqualini 1 , Francesca Bergomas 1 , Samantha Pesce 1 , Marina Sironi 1 , Fabio Grizzi 1 , Alberto Mantovani 1, 2 , Cristina Belgiovine 1, * , Paola Allavena 1, 2, * 1 Department of Immunology and Inflammation, IRCCS-Humanitas Clinical and Research Center, 20089 Rozzano, Milano, Italy 2 Humanitas University, 20089 Rozzano, Milano, Italy * These authors have contributed equally to this work Correspondence to: Cristina Belgiovine, email: cristina.belgiovine@humanitasresearch.it Paola Allavena, email: paola.allavena@humanitasresearch.it Keywords: TRAIL, TRAIL receptors, apoptosis, tumor-associated macrophages, targeting macrophages Received: February 17, 2016 Accepted: April 06, 2016 Published: May 13, 2016 ABSTRACT Despite the accepted dogma that TRAIL kills only tumor cells and spares normal ones, we show in this study that mononuclear phagocytes are susceptible to recombinant TRAIL via caspase-dependent apoptosis. Human resting monocytes and in vitro -differentiated macrophages expressed substantial levels of the functional TRAIL receptors (TRAIL-R1 and TRAIL-R2), while neutrophils and lymphocytes mostly expressed the non-signaling decoy receptor (TRAIL-R3). Accordingly, exclusively monocytes and macrophages activated caspase-8 and underwent apoptosis upon recombinant TRAIL treatment. TRAIL-Rs were up-regulated by anti-inflammatory agents (IL-10, glucocorticoids) and by natural compounds (Apigenin, Quercetin, Palmitate) and their treatment resulted in increased TRAIL-induced apoptosis. In mice, the only signaling TRAIL-R (DR5) was preferentially expressed by blood monocytes rather than neutrophils or lymphocytes. In both mice and humans, Tumor-Associated Macrophages (TAM) expressed functional TRAIL-R, while resident macrophages in normal tissues did not. As a proof of principle, we treated mice bearing a murine TRAIL-resistant fibrosarcoma with recombinant TRAIL. We observed significant decrease of circulating monocytes and infiltrating TAM, as well as reduced tumor growth and lower metastasis formation. Overall, these findings demonstrate that human and murine monocytes/macrophages are, among leukocytes, uniquely susceptible to TRAIL-mediated killing. This differential susceptibility to TRAIL could be exploited to selectively target macrophages in tumors.
46. The CCL3 family of chemokines and innate immunity cooperate in vivo in the eradication of an established lymphoma xenograft by rituximab
- Author
-
Martino Introna, Elena Cittera, Fabio Pasqualini, Marzia Leidi, Josée Golay, Annunciata Vecchi, Chiara Buracchi, Silvano Sozzani, and J. Douglas Waterfield
- Subjects
Male ,Chemokine ,Transplantation, Heterologous ,Immunology ,Mice, Nude ,Antibodies, Monoclonal, Murine-Derived ,Mice ,Immune system ,immune system diseases ,In vivo ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Immunology and Allergy ,RNA, Messenger ,Chemokine CCL4 ,Chemokine CCL3 ,CD20 ,Innate immune system ,biology ,Antibodies, Monoclonal ,Complement System Proteins ,medicine.disease ,Burkitt Lymphoma ,Immunity, Innate ,Lymphoma ,Gene Expression Regulation, Neoplastic ,Transplantation ,Chemokines, CC ,Multigene Family ,biology.protein ,Rituximab ,medicine.drug - Abstract
The therapeutic mAb rituximab induced the expression of the CCL3 and CCL4 chemokines in the human lymphoma line BJAB following binding to the CD20 Ag. Induction of CCL3/4 in vitro was specific, was observed in several cell lines and freshly isolated lymphoma samples and also took place at the protein level in vitro and in vivo. To investigate the role of these β-chemokines in the mechanism of action of rituximab, we synthesized a N-terminally truncated CCL3 molecule CCL3(11–70), which had antagonist activity on chemotaxis mediated by either CCL3 or BJAB supernatant. We also set up an established s.c. BJAB tumor model in athymic mice. Rituximab, given weekly after tumors had reached 250 mm2, led to complete disappearance of the lymphoma within 2–3 wk. Treatment of mice with cobra venom factor showed that complement was required for rituximab therapeutic activity. Treatment of BJAB tumor bearing mice every 2 days with the CCL3(11–70) antagonist, starting 1 wk before rituximab treatment, had no effect on tumor growth by itself, but completely inhibited the therapeutic activity of the Ab. To determine whether CCL3 acts through recruitment/activation of immune cells, we specifically depleted NK cells, polymorphonuclear cells, and macrophages using mAbs, clodronate treatment, or Rag2−/−cγ−/− mice. The data demonstrated that these different cell populations are involved in BJAB tumor eradication. We propose that rituximab rapidly activates complement and induces β-chemokines in vivo, which in turn activate the innate immunity network required for efficient eradication of the bulky BJAB tumor.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.