Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alessandro Aiuti is active.

Publication


Featured researches published by Alessandro Aiuti.


The New England Journal of Medicine | 2009

Gene Therapy for immunodeficiency due to Adenosine Deaminase Deficiency

Alessandro Aiuti; Federica Cattaneo; Stefania Galimberti; Ulrike Benninghoff; Barbara Cassani; Luciano Callegaro; Samantha Scaramuzza; Grazia Andolfi; Massimiliano Mirolo; Immacolata Brigida; Antonella Tabucchi; Filippo Carlucci; Martha M. Eibl; Memet Aker; Shimon Slavin; Hamoud Al-Mousa; Abdulaziz Al Ghonaium; Alina Ferster; Andrea Duppenthaler; Luigi D. Notarangelo; Uwe Wintergerst; Rebecca H. Buckley; Marco Bregni; Sarah Marktel; Maria Grazia Valsecchi; Pier Luca Rossi; Fabio Ciceri; Miniero R; Claudio Bordignon; Maria Grazia Roncarolo

BACKGROUND We investigated the long-term outcome of gene therapy for severe combined immunodeficiency (SCID) due to the lack of adenosine deaminase (ADA), a fatal disorder of purine metabolism and immunodeficiency. METHODS We infused autologous CD34+ bone marrow cells transduced with a retroviral vector containing the ADA gene into 10 children with SCID due to ADA deficiency who lacked an HLA-identical sibling donor, after nonmyeloablative conditioning with busulfan. Enzyme-replacement therapy was not given after infusion of the cells. RESULTS All patients are alive after a median follow-up of 4.0 years (range, 1.8 to 8.0). Transduced hematopoietic stem cells have stably engrafted and differentiated into myeloid cells containing ADA (mean range at 1 year in bone marrow lineages, 3.5 to 8.9%) and lymphoid cells (mean range in peripheral blood, 52.4 to 88.0%). Eight patients do not require enzyme-replacement therapy, their blood cells continue to express ADA, and they have no signs of defective detoxification of purine metabolites. Nine patients had immune reconstitution with increases in T-cell counts (median count at 3 years, 1.07x10(9) per liter) and normalization of T-cell function. In the five patients in whom intravenous immune globulin replacement was discontinued, antigen-specific antibody responses were elicited after exposure to vaccines or viral antigens. Effective protection against infections and improvement in physical development made a normal lifestyle possible. Serious adverse events included prolonged neutropenia (in two patients), hypertension (in one), central-venous-catheter-related infections (in two), Epstein-Barr virus reactivation (in one), and autoimmune hepatitis (in one). CONCLUSIONS Gene therapy, combined with reduced-intensity conditioning, is a safe and effective treatment for SCID in patients with ADA deficiency. (ClinicalTrials.gov numbers, NCT00598481 and NCT00599781.)


Journal of Clinical Investigation | 2007

Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy

Alessandro Aiuti; Barbara Cassani; Grazia Andolfi; Massimiliano Mirolo; Luca Biasco; Fabrizia Urbinati; Cristina Valacca; Samantha Scaramuzza; Memet Aker; Shimon Slavin; Matteo Cazzola; Daniela Sartori; Alessandro Ambrosi; Clelia Di Serio; Maria Grazia Roncarolo; Fulvio Mavilio; Claudio Bordignon

Gene transfer into HSCs is an effective treatment for SCID, although potentially limited by the risk of insertional mutagenesis. We performed a genome-wide analysis of retroviral vector integrations in genetically corrected HSCs and their multilineage progeny before and up to 47 months after transplantation into 5 patients with adenosine deaminase-deficient SCID. Gene-dense regions, promoters, and transcriptionally active genes were preferred retroviral integrations sites (RISs) both in preinfusion transduced CD34(+) cells and in vivo after gene therapy. The occurrence of insertion sites proximal to protooncogenes or genes controlling cell growth and self renewal, including LMO2, was not associated with clonal selection or expansion in vivo. Clonal analysis of long-term repopulating cell progeny in vivo revealed highly polyclonal T cell populations and shared RISs among multiple lineages, demonstrating the engraftment of multipotent HSCs. These data have important implications for the biology of retroviral vectors, the dynamics of genetically modified HSCs, and the safety of gene therapy.


European Journal of Immunology | 1999

EXPRESSION OF CXCR4, THE RECEPTOR FOR STROMAL CELL-DERIVED FACTOR-1 ON FETAL AND ADULT HUMAN LYMPHOHEMATOPOIETIC PROGENITORS

Alessandro Aiuti; Manuela Tavian; Arcadi Cipponi; Francesca Ficara; Elisabetta Zappone; James A. Hoxie; Bruno Péault; Claudio Bordignon

Stromal cell‐derived factor‐1 (SDF‐1) is a CXC chemokine produced by stromal cells that acts as a chemoattractant for human CD34+ progenitor cells. We investigated the expression of CXCR4, the receptor for SDF‐1, on CD34+ cells from different hematopoietic sites and developmental stages. CXCR4 was detected by flow cytometry on 37 % of fetal bone marrow (BM) [gestation weeks (gw) 14 – 23] and 40 % of adult BM CD34+ cells. Interestingly, in fetal liver CD34+ cells, CXCR4 was expressed at lower levels at later stages (9 %, gw 20 – 23) compared to early stages of development (39 %, gw 7.5 – 18), suggesting a development‐related change in the migratory capacity of progenitors. CXCR4 was detected at similar levels on both phenotypically primitive and committed progenitors from fetal and adult sites. However, B cell lineage progenitor and precursor cells expressed CXCR4 at the highest density (80 % of BM CD34+/CD10+ pro‐B cells are CXCR4+). CXCR4 was also expressed in the fetal thymus in early T cell precursors and found to be down‐regulated during T cell maturation. Finally, we found that stem cell factor, alone or in combination with other cytokines, can up‐modulate CXCR4 expression on CD34+ cells by three‐ to fourfold. In conclusion, our results suggest that CXCR4 may play an important role in the local and systemic trafficking of human CD34+ cells as well as in human B lymphopoiesis and that its expression can be modulated by cytokines.


Immunity | 2002

Wiskott-Aldrich Syndrome Protein Regulates Lipid Raft Dynamics during Immunological Synapse Formation

Loı̈c Dupré; Alessandro Aiuti; Sara Trifari; Silvana Martino; Paola Saracco; Claudio Bordignon; Maria Grazia Roncarolo

Immunological synapse assembly relies on the clustering of lipid rafts and is required for optimal T cell activation. We demonstrate that the Wiskott-Aldrich syndrome protein (WASP) is recruited to lipid rafts immediately after TCR and CD28 triggering and is required for the movements of lipid rafts. T cells from Wiskott-Aldrich syndrome (WAS) patients, lacking WASP, proliferate poorly after TCR/CD28 activation and have impaired capacities to cluster the lipid raft marker GM1 and to upregulate GM1 cell surface expression. T cell proliferation and lipid raft clustering are restored by retroviral transfer of the WASP gene. These results demonstrate that WASP plays a central role in the movements of lipid rafts and identify a potential mechanism underlying the T cell defect affecting WAS patients.


Blood | 2009

How I treat ADA deficiency

H. Bobby Gaspar; Alessandro Aiuti; Fulvio Porta; Fabio Candotti; Michael S. Hershfield; Luigi D. Notarangelo

Adenosine deaminase deficiency is a disorder of purine metabolism leading to severe combined immunodeficiency (ADA-SCID). Without treatment, the condition is fatal and requires early intervention. Haematopoietic stem cell transplantation is the major treatment for ADA-SCID, although survival following different donor sources varies considerably. Unlike other SCID forms, 2 other options are available for ADA-SCID: enzyme replacement therapy (ERT) with pegylated bovine ADA, and autologous haematopoietic stem cell gene therapy (GT). Due to the rarity of the condition, the lack of large scale outcome studies, and availability of different treatments, guidance on treatment strategies is limited. We have reviewed the currently available evidence and together with our experience of managing this condition propose a consensus management strategy. Matched sibling donor transplants represent a successful treatment option with high survival rates and excellent immune recovery. Mismatched parental donor transplants have a poor survival outcome and should be avoided unless other treatments are unavailable. ERT and GT both show excellent survival, and therefore the choice between ERT, MUD transplant, or GT is difficult and dependent on several factors, including accessibility to the different modalities, response of patients to long-term ERT, and the attitudes of physicians and parents to the short- and potential long-term risks associated with different treatments.


Nature Medicine | 2003

Safety of retroviral gene marking with a truncated NGF receptor

Chiara Bonini; Manuel Grez; Catia Traversari; Fabio Ciceri; S. Marktel; Giuliana Ferrari; Mary C. Dinauer; Mohammed A. Sadat; Alessandro Aiuti; S. Deola; M. Radrizzani; Anton Hagenbeek; J. Apperley; S. Ebeling; A. Martens; H. J. Kolb; M. Weber; Francesco Lotti; Alexis Grande; E. Weissinger; Juan A. Bueren; M. Lamana; J. H.F. Falkenburg; M. H.M. Heemskerk; T. Austin; Steven M. Kornblau; F. Marini; C. Benati; Zulma Magnani; S. Cazzaniga

To the editor—Random integration into the host cell genome and inappropriate transgene expression are major safety concerns for the clinical use of retroviral vectors. Li et al. recently reported a leukemic transformation of mouse bone marrow cells caused by integration of a transgene-carrying retroviral vector into the Evi1 proto-oncogene. They suggested that expression of the transgene, a truncated form of the p75 low-affinity nerve growth factor receptor (∆LNGFR) with most of the intracytoplasmic tail deleted (from residue 248), contributed to the leukemic progression. Because ∆LNGFR is used as a surface marker in gene therapy clinical trials aimed at controlling graft-versus-host disease (GVHD) after bone marrow transplantation (BMT), a critical assessment of the potential risks associated with the use of such a molecule is essential. In a collaborative effort between 17 independent groups of investigators, we have accumulated both pre-clinical and clinical evidence supporting the safety of ∆LNGFR as a cell-marking molecule. Cumulative data obtained from >300 mice transplanted with bone marrow cells transduced with ∆LNGFR-expressing retroviral vectors showed normal engraftment, persistence and differentiation of ∆LNGFR-expressing hematopoietic stemprogenitor cells (HSCs) in primary, secondary and tertiary BMT recipients, with no adverse events (Table 1 and Supplementary Information online). Over 100 of these mice were monitored for >20 weeks after BMT; more than 70 animals, including 16 recipients of secondary or tertiary BMT, were monitored for >28 weeks. Considering that a total of >1 × 10 transduced cells were transplanted, and assuming an average of one retroviral integration per cell, we estimate the risk of oncogenic transformation after transduction with a ∆LNGFR-encoding retroviral vector to be <1 in 10 integration events. Therefore, expression of ∆LNGFR could not have increased the expected frequency of an insertional oncogenesis event, which has been previously estimated at 10 to 10 per insertion event. Expression of ∆LNGFR did not alter the function or survival of T lymphocytes derived from peripheral blood mononuclear cells transduced with a variety of vectors and studied in different animal models. In pre-clinical models of post-BMT GVHD, no difference in the ability to induce donor chimerism or to mediate GVHD was observed for ∆LNGFR-expressing T cells, as compared with control T cells, in 356 mice, 200 rats and 3 dogs (Table 1 and Supplementary Information online), again with no adverse events. Analysis of 102 independent transductions of human peripheral lymphocytes with two different vectors (SFCMM-3 and SFCM) encoding the same ∆LNGFR detected no change in the expression of markers of lineage, activation or adhesion, or in the proliferative capacity of T cells, as assayed by limiting dilution after polyclonal in vitro stimulation. All cells remained strictly dependent on interleukin-2 for growth and survival, and the Safety of retroviral gene marking with a truncated NGF receptor


Blood | 2009

Recent advances in understanding the pathophysiology of Wiskott-Aldrich syndrome

Marita Bosticardo; Francesco Marangoni; Alessandro Aiuti; Anna Villa; Maria Grazia Roncarolo

Wiskott-Aldrich syndrome (WAS) is a severe X-linked immunodeficiency caused by mutations in the gene encoding for WASP, a key regulator of signaling and cytoskeletal reorganization in hematopoietic cells. Mutations in WASP result in a wide spectrum of clinical manifestations ranging from the relatively mild X-linked thrombocytopenia to the classic full-blown WAS phenotype characterized by thrombocytopenia, immunodeficiency, eczema, and high susceptibility to developing tumors and autoimmune manifestations. The life expectancy of patients affected by severe WAS is reduced, unless they are successfully cured by bone marrow transplantation from related identical or matched unrelated donors. Because many patients lack a compatible bone marrow donor, the administration of WAS gene-corrected autologous hematopoietic stem cells could represent an alternative therapeutic approach. In the present review, we focus on recent progress in understanding the molecular and cellular mechanisms contributing to the pathophysiology of WAS. Although molecular and cellular studies have extensively analyzed the mechanisms leading to defects in T, B, and dendritic cells, the basis of autoimmunity and thrombocytopenia still remains poorly understood. A full understanding of these mechanisms is still needed to further implement new therapeutic strategies for this peculiar immunodeficiency.


Journal of Experimental Medicine | 2007

WASP regulates suppressor activity of human and murine CD4(+)CD25(+)FOXP3(+) natural regulatory T cells.

Francesco Marangoni; Sara Trifari; Samantha Scaramuzza; Cristina Panaroni; Silvana Martino; Luigi D. Notarangelo; Zeina Baz; Ayse Metin; Federica Cattaneo; Anna Villa; Alessandro Aiuti; Manuela Battaglia; Maria Grazia Roncarolo; Loïc Dupré

A large proportion of Wiskott-Aldrich syndrome (WAS) patients develop autoimmunity and allergy. CD4+CD25+FOXP3+ natural regulatory T (nTreg) cells play a key role in peripheral tolerance to prevent immune responses to self-antigens and allergens. Therefore, we investigated the effect of WAS protein (WASP) deficiency on the distribution and suppressor function of nTreg cells. In WAS−/− mice, the steady-state distribution and phenotype of nTreg cells in the thymus and spleen were normal. However, WAS−/− nTreg cells engrafted poorly in immunized mice, indicating perturbed homeostasis. Moreover, WAS−/− nTreg cells failed to proliferate and to produce transforming growth factor β upon T cell receptor (TCR)/CD28 triggering. WASP-dependent F-actin polarization to the site of TCR triggering might not be involved in WAS−/− nTreg cell defects because this process was also inefficient in wild-type (WT) nTreg cells. Compared with WT nTreg cells, WAS−/− nTreg cells showed reduced in vitro suppressor activity on both WT and WAS−/− effector T cells. Similarly, peripheral nTreg cells were present at normal levels in WAS patients but failed to suppress proliferation of autologous and allogeneic CD4+ effector T cells in vitro. Thus, WASP appears to play an important role in the activation and suppressor function of nTreg cells, and a dysfunction or incorrect localization of nTreg cells may contribute to the development of autoimmunity in WAS patients.


Nature Medicine | 2009

Comprehensive genomic access to vector integration in clinical gene therapy.

Richard Gabriel; Ralph Eckenberg; Anna Paruzynski; Cynthia C. Bartholomae; Ali Nowrouzi; Anne Arens; Steven J. Howe; Claudia Cattoglio; Wei Wang; Katrin Faber; Kerstin Schwarzwaelder; Romy Kirsten; Annette Deichmann; Claudia R. Ball; Kamaljit S. Balaggan; Rafael J. Yáñez-Muñoz; Robin R. Ali; H. Bobby Gaspar; Luca Biasco; Alessandro Aiuti; Daniela Cesana; Eugenio Montini; Luigi Naldini; Odile Cohen-Haguenauer; Fulvio Mavilio; Aj Thrasher; Hanno Glimm; Christof von Kalle; William Saurin; Manfred Schmidt

Retroviral vectors have induced subtle clonal skewing in many gene therapy patients and severe clonal proliferation and leukemia in some of them, emphasizing the need for comprehensive integration site analyses to assess the biosafety and genomic pharmacokinetics of vectors and clonal fate of gene-modified cells in vivo. Integration site analyses such as linear amplification–mediated PCR (LAM-PCR) require a restriction digest generating unevenly small fragments of the genome. Here we show that each restriction motif allows for identification of only a fraction of all genomic integrants, hampering the understanding and prediction of biological consequences after vector insertion. We developed a model to define genomic access to the viral integration site that provides optimal restriction motif combinations and minimizes the percentage of nonaccessible insertion loci. We introduce a new nonrestrictive LAM-PCR approach that has superior capabilities for comprehensive unbiased integration site retrieval in preclinical and clinical samples independent of restriction motifs and amplification inefficiency.


Current Opinion in Immunology | 2012

Gene therapy for primary immunodeficiencies: Part 2

Marina Cavazzana-Calvo; Alain Fischer; Salima Hacein-Bey-Abina; Alessandro Aiuti

Over 60 patients affected by SCID due to IL2RG deficiency (SCID-X1) or adenosine deaminase (ADA)-SCID have received hematopoietic stem cell gene therapy in the past 15 years using gammaretroviral vectors, resulting in immune reconstitution and clinical benefit in the majority of them. However, the occurrence of insertional oncogenesis in the SCID-X1 trials has led to the development of new clinical trials based on integrating vectors with improved safety design as well as investigation on new technologies for highly efficient gene targeting and site-specific gene editing. Here we will present the experience and perspectives of gene therapy for SCID-X1 and ADA-SCID and discuss the pros and cons of gene therapy in comparison to allogeneic transplantation.

Collaboration


Dive into the Alessandro Aiuti's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samantha Scaramuzza

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Luigi Naldini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Immacolata Brigida

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Fabio Ciceri

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Anna Villa

National Research Council

View shared research outputs
Top Co-Authors

Avatar

Luca Biasco

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Maria Pia Cicalese

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Marita Bosticardo

Vita-Salute San Raffaele University

View shared research outputs
Researchain Logo
Decentralizing Knowledge