Network


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

Hotspot


Dive into the research topics where Alessio Cantore is active.

Publication


Featured researches published by Alessio Cantore.


Nature Methods | 2011

Site-specific integration and tailoring of cassette design for sustainable gene transfer

Angelo Lombardo; Daniela Cesana; Pietro Genovese; Bruno Di Stefano; Elena Provasi; Daniele F Colombo; Margherita Neri; Zulma Magnani; Alessio Cantore; Pietro Lo Riso; Martina Damo; Oscar M Pello; Michael C. Holmes; Philip D. Gregory; Angela Gritti; Vania Broccoli; Chiara Bonini; Luigi Naldini

Integrative gene transfer methods are limited by variable transgene expression and by the consequences of random insertional mutagenesis that confound interpretation in gene-function studies and may cause adverse events in gene therapy. Site-specific integration may overcome these hurdles. Toward this goal, we studied the transcriptional and epigenetic impact of different transgene expression cassettes, targeted by engineered zinc-finger nucleases to the CCR5 and AAVS1 genomic loci of human cells. Analyses performed before and after integration defined features of the locus and cassette design that together allow robust transgene expression without detectable transcriptional perturbation of the targeted locus and its flanking genes in many cell types, including primary human lymphocytes. We thus provide a framework for sustainable gene transfer in AAVS1 that can be used for dependable genetic manipulation, neutral marking of the cell and improved safety of therapeutic applications, and demonstrate its feasibility by rapidly generating human lymphocytes and stem cells carrying targeted and benign transgene insertions.


Hepatology | 2011

Hepatocyte-targeted expression by integrase-defective lentiviral vectors induces antigen-specific tolerance in mice with low genotoxic risk

Janka Matrai; Alessio Cantore; Cynthia C. Bartholomae; Andrea Annoni; Wei Wang; Abel Acosta-Sanchez; Ermira Samara-Kuko; Liesbeth De Waele; Ling Ma; Pietro Genovese; Martina Damo; Anne Arens; Kevin S. Goudy; Timothy C. Nichols; Christof von Kalle; Marinee Chuah; Maria Grazia Roncarolo; Manfred Schmidt; Thierry Vandendriessche; Luigi Naldini

Lentiviral vectors are attractive tools for liver‐directed gene therapy because of their capacity for stable gene expression and the lack of preexisting immunity in most human subjects. However, the use of integrating vectors may raise some concerns about the potential risk of insertional mutagenesis. Here we investigated liver gene transfer by integrase‐defective lentiviral vectors (IDLVs) containing an inactivating mutation in the integrase (D64V). Hepatocyte‐targeted expression using IDLVs resulted in the sustained and robust induction of immune tolerance to both intracellular and secreted proteins, despite the reduced transgene expression levels in comparison with their integrase‐competent vector counterparts. IDLV‐mediated and hepatocyte‐targeted coagulation factor IX (FIX) expression prevented the induction of neutralizing antibodies to FIX even after antigen rechallenge in hemophilia B mice and accounted for relatively prolonged therapeutic FIX expression levels. Upon the delivery of intracellular model antigens, hepatocyte‐targeted IDLVs induced transgene‐specific regulatory T cells that contributed to the observed immune tolerance. Deep sequencing of IDLV‐transduced livers showed only rare genomic integrations that had no preference for gene coding regions and occurred mostly by a mechanism inconsistent with residual integrase activity. Conclusion: IDLVs provide an attractive platform for the tolerogenic expression of intracellular or secreted proteins in the liver with a substantially reduced risk of insertional mutagenesis. (HEPATOLOGY 2011;)


Blood | 2009

In vivo delivery of a microRNA-regulated transgene induces antigen-specific regulatory T cells and promotes immunologic tolerance

Andrea Annoni; Brian D. Brown; Alessio Cantore; Lucia Sergi Sergi; Luigi Naldini; Maria Grazia Roncarolo

We previously showed that incorporating target sequences for the hematopoietic-specific microRNA miR-142 into an antigen-encoding transgene prevents antigen expression in antigen-presenting cells (APCs). To determine whether this approach induces immunologic tolerance, we treated mice with a miR-142-regulated lentiviral vector encoding green fluorescent protein (GFP), and subsequently vaccinated the mice against GFP. In contrast to control mice, no anti-GFP response was observed, indicating that robust tolerance to the transgene-encoded antigen was achieved. Furthermore, injection of the miR-142-regulated vector induced a population of GFP-specific regulatory T cells. Interestingly, an anti-GFP response was observed when microRNA miR-122a was inserted into the vector and antigen expression was detargeted from hepatocytes as well as APCs. This demonstrates that, in the context of lentiviral vector-mediated gene transfer, detargeting antigen expression from professional APCs, coupled with expression in hepatocytes, can induce antigen-specific immunologic tolerance.


Blood | 2012

Hyperfunctional coagulation factor IX improves the efficacy of gene therapy in hemophilic mice

Alessio Cantore; Nisha Nair; Patrizia Della Valle; Mario Di Matteo; Janka Matrai; Francesca Sanvito; Chiara Brombin; Clelia Di Serio; Armando D'Angelo; Marinee Chuah; Luigi Naldini; Thierry Vandendriessche

Gene therapy may provide a cure for hemophilia and overcome the limitations of protein replacement therapy. Increasing the potency of gene transfer vectors may allow improvement of their therapeutic index, as lower doses can be administered to achieve therapeutic benefit, reducing toxicity of in vivo administration. Here we generated codon-usage optimized and hyperfunctional factor IX (FIX) transgenes carrying an R338L amino acid substitution (FIX Padua), previously associated with clotting hyperactivity and thrombophilia. We delivered these transgenes to hemophilia B mice by hepatocyte-targeted integration-competent and -defective lentiviral vectors. The hyperfunctional FIX transgenes increased FIX activity reconstituted in the plasma without detectable adverse effects, allowing correction of the disease phenotype at lower vector doses and resulting in improved hemostasis in vivo. The combined effect of codon optimization with the hyperactivating FIX-R338L mutation resulted in a robust 15-fold gain in potency and therefore provides a promising strategy to improve the efficacy, feasibility, and safety of hemophilia gene therapy.


Science Translational Medicine | 2015

Liver-directed lentiviral gene therapy in a dog model of hemophilia B

Alessio Cantore; Marco Ranzani; Cynthia C. Bartholomae; Monica Volpin; Patrizia Della Valle; Francesca Sanvito; Lucia Sergi Sergi; Pierangela Gallina; Fabrizio Benedicenti; Dwight A. Bellinger; Robin A. Raymer; Elizabeth P. Merricks; Francesca Bellintani; Samia Martin; Claudio Doglioni; Armando D'Angelo; Thierry Vandendriessche; Marinee Chuah; Manfred Schmidt; Timothy C. Nichols; Eugenio Montini; Luigi Naldini

Gene therapy with lentiviral vectors targeting transgene expression to hepatocytes provides stable reconstitution of clotting activity in dogs with hemophilia B and does not show genotoxicity in tumor-prone mice. Advancing gene therapy for hemophilia Hemophilia is an inherited bleeding disorder caused by a deficiency in a blood clotting factor. The current treatment requires lifelong intravenous administration of the missing clotting factor every few days, a costly and demanding regimen for patients with hemophilia. Gene therapy has the potential to provide a single-shot treatment option by replacing a functional gene in liver cells that naturally produce the factor. Cantore et al. now report a study of the efficacy and safety of liver-directed in vivo gene therapy in large and small animal models using lentiviral vectors. This gene therapy strategy with lentiviral vectors may complement the use of other gene therapy vectors for treating hemophilia. We investigated the efficacy of liver-directed gene therapy using lentiviral vectors in a large animal model of hemophilia B and evaluated the risk of insertional mutagenesis in tumor-prone mouse models. We showed that gene therapy using lentiviral vectors targeting the expression of a canine factor IX transgene in hepatocytes was well tolerated and provided a stable long-term production of coagulation factor IX in dogs with hemophilia B. By exploiting three different mouse models designed to amplify the consequences of insertional mutagenesis, we showed that no genotoxicity was detected with these lentiviral vectors. Our findings suggest that lentiviral vectors may be an attractive candidate for gene therapy targeted to the liver and may be potentially useful for the treatment of hemophilia.


Embo Molecular Medicine | 2013

Liver gene therapy by lentiviral vectors reverses anti-factor IX pre-existing immunity in haemophilic mice.

Andrea Annoni; Alessio Cantore; Patrizia Della Valle; Kevin S. Goudy; Mahzad Akbarpour; Fabio Russo; Sara Bartolaccini; Armando D'Angelo; Maria Grazia Roncarolo; Luigi Naldini

A major complication of factor replacement therapy for haemophilia is the development of anti‐factor neutralizing antibodies (inhibitors). Here we show that liver gene therapy by lentiviral vectors (LVs) expressing factor IX (FIX) strongly reduces pre‐existing anti‐FIX antibodies and eradicates FIX inhibitors in haemophilia B mice. Concomitantly, plasma FIX levels and clotting activity rose to 50–100% of normal. The treatment was effective in 75% of treated mice. FIX‐specific plasma cells (PCs) and memory B cells were reduced, likely because of memory B‐cell depletion in response to constant exposure to high doses of FIX. Regulatory T cells displaying FIX‐specific suppressive capacity were induced in gene therapy treated mice and controlled FIX‐specific T helper cells. Gene therapy proved safer than a regimen mimicking immune tolerance induction (ITI) by repeated high‐dose FIX protein administration, which induced severe anaphylactoid reactions in inhibitors‐positive haemophilia B mice. Liver gene therapy can thus reverse pre‐existing immunity, induce active tolerance to FIX and establish sustained FIX activity at therapeutic levels. These data position gene therapy as an attractive treatment option for inhibitors‐positive haemophilic patients.


Science Translational Medicine | 2015

Insulin B chain 9–23 gene transfer to hepatocytes protects from type 1 diabetes by inducing Ag-specific FoxP3+ Tregs

Mahzad Akbarpour; Kevin S. Goudy; Alessio Cantore; Fabio Russo; Francesca Sanvito; Luigi Naldini; Andrea Annoni; Maria Grazia Roncarolo

Hepatocyte-targeted InsB9–23 gene transfer protects from type 1 diabetes by inducing antigen-specific regulatory T cells. Gene therapy for diabetes Gene therapy is being used with increasing success to treat a rapidly growing group of diseases ranging from monogenetic diseases to cancer. Now, Akbarpour et al. suggest that gene transfer may help protect against type 1 diabetes.The authors used a lentiviral vector to express insulin in liver cells in a mouse model of type 1 diabetes. This therapy induced regulatory T cell specific for insulin, but not a control antigen, and halted immune cell infiltration into the pancreatic islet. Moreover, when this therapy was combined with a single dose of anti-CD3 monoclonal antibody, it stopped disease progression in these mice. These data suggest that expressing autoantigen in liver cells may induce antigen-specific tolerance in autoimmune disease. Antigen (Ag)–specific tolerance in type 1 diabetes (T1D) in human has not been achieved yet. Targeting lentiviral vector (LV)–mediated gene expression to hepatocytes induces active tolerance toward the encoded Ag. The insulin B chain 9–23 (InsB9–23) is an immunodominant T cell epitope in nonobese diabetic (NOD) mice. To determine whether auto-Ag gene transfer to hepatocytes induces tolerance and control of T1D, NOD mice were treated with integrase-competent LVs (ICLVs) that selectively target the expression of InsB9–23 to hepatocytes. ICLV treatment induced InsB9–23–specific effector T cells but also FoxP3+ regulatory T cells (Tregs), which halted islet immune cell infiltration, and protected from T1D. Moreover, ICLV treatment combined with a single suboptimal dose of anti-CD3 monoclonal antibody (mAb) is effective in T1D reversal. Splenocytes from LV.InsB9–23–treated mice, but not from LV.OVA (ovalbumin)–treated control mice, stopped diabetes development, demonstrating that protection is Ag-specific. Depletion of CD4+CD25+FoxP3+ T cells led to diabetes progression, indicating that Ag-specific FoxP3+ Tregs mediate protection. Integrase-defective LVs (IDLVs).InsB9–23, which alleviate the concerns for insertional mutagenesis and support transient transgene expression in hepatocytes, were also efficient in protecting from T1D. These data demonstrate that hepatocyte-targeted auto-Ag gene expression prevents and resolves T1D and that stable integration of the transgene is not required for this protection. Gene transfer to hepatocytes can be used to induce Ag-specific tolerance in autoimmune diseases.


Embo Molecular Medicine | 2017

Genome editing for scalable production of alloantigen‐free lentiviral vectors for in vivo gene therapy

Michela Milani; Andrea Annoni; Sara Bartolaccini; Mauro Biffi; Fabio Russo; Tiziano Di Tomaso; Andrea Raimondi; Johannes Lengler; Michael C. Holmes; Friedrich Scheiflinger; Angelo Lombardo; Alessio Cantore; Luigi Naldini

Lentiviral vectors (LV) are powerful and versatile vehicles for gene therapy. However, their complex biological composition challenges large‐scale manufacturing and raises concerns for in vivo applications, because particle components and contaminants may trigger immune responses. Here, we show that producer cell‐derived polymorphic class‐I major histocompatibility complexes (MHC‐I) are incorporated into the LV surface and trigger allogeneic T‐cell responses. By disrupting the beta‐2 microglobulin gene in producer cells, we obtained MHC‐free LV with substantially reduced immunogenicity. We introduce this targeted editing into a novel stable LV packaging cell line, carrying single‐copy inducible vector components, which can be reproducibly converted into high‐yield LV producers upon site‐specific integration of the LV genome of interest. These LV efficiently transfer genes into relevant targets and are more resistant to complement‐mediated inactivation, because of reduced content of the vesicular stomatitis virus envelope glycoprotein G compared to vectors produced by transient transfection. Altogether, these advances support scalable manufacturing of alloantigen‐free LV with higher purity and increased complement resistance that are better suited for in vivo gene therapy.


Cellular Immunology | 2018

Modulation of immune responses in lentiviral vector-mediated gene transfer

Andrea Annoni; Silvia Gregori; Luigi Naldini; Alessio Cantore

Highlights • Immune responses may be detrimental to both the safety and efficacy of gene therapy.• Design and manufacturing can be tuned to reduce vector immunogenicity.• Active transgene-specific immune tolerance is desirable in gene therapy.• Novel targeted immune-modulatory strategies can be explored to improve gene therapy.


Molecular Therapy | 2016

286. Genome Editing of Inducible Cell Lines for Scalable Production of Improved Lentiviral Vectors for Human Gene Therapy

Alessio Cantore; Michela Milani; Andrea Annoni; Johannes Lengler; Sara Bartolaccini; Tiziano Di Tomaso; Michael C. Holmes; Friedrich Scheiflinger; Angelo Lombardo; Luigi Naldini

Lentiviral vectors (LVs) represent efficient and versatile vehicles for gene therapy. Current manufacturing of clinical-grade LVs mostly relies on transient transfection of plasmids expressing the multiple vector components. This method is labor and cost intensive and becomes challenging when facing the need of scale-up and standardization. The development of stable LV producer cell lines will greatly facilitate overcoming these hurdles. We have generated an inducible LV packaging cell line, carrying the genes encoding for third-generation vector components stably integrated in the genome under the control of tetracycline-regulated promoters. These LV packaging cells are stable in culture even after single-cell cloning and can be scaled up to large volumes. In order to minimize the immunogenicity of LVs for in vivo administration, we set out to remove the highly polymorphic class-I major histocompatibility complexes (MHC-I) expressed on LV packaging cells and incorporated in the LV envelope. We performed genetic disruption of the β-2 microglobulin (B2M) gene, a required component for the assembly and trafficking of all MHC-I to the plasma membrane in LV producer cells, exploiting the RNA-guided Cas9 nuclease. The resulting B2M-negative cells were devoid of surface-exposed MHC-I and produced MHC-free LVs. These LVs retain their infectivity on all tested cells in vitro and efficiently transduced the mouse liver upon intravenous administration. Strikingly, the MHC-free LVs showed significantly reduced immunogenicity in a T-cell activation assay performed on human primary T cells co-cultured with syngeneic monocytes exposed to LV, from several (n=7) healthy donors. To reproducibly generate LV-producer cell lines from these cells, we insert the LV genome of interest in the AAVS1 locus, chosen for robust expression, exploiting engineered nucleases and homology-directed repair. By this strategy, we have obtained several independent producer cell lines for LVs that express marker or therapeutic genes and are devoid of plasmid DNA contamination. LVs produced by these cells reproducibly show titer and infectivity within the lower bound range of standard optimized transient transfection, and effectively transduce relevant target cells, such as hematopoietic stem/progenitor cells and T cells ex vivo and the mouse liver in vivo. Overall, we provide evidence that rationally designed targeted genome engineering can be used to improve the yield, quality, safety and sustainability of LV production for clinical use.

Collaboration


Dive into the Alessio Cantore's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea Annoni

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Armando D'Angelo

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Timothy C. Nichols

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Marinee Chuah

Free University of Brussels

View shared research outputs
Top Co-Authors

Avatar

Patrizia Della Valle

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Lucia Sergi Sergi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Angelo Lombardo

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Dwight A. Bellinger

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge