Network


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

Hotspot


Dive into the research topics where Marina Cavazzana is active.

Publication


Featured researches published by Marina Cavazzana.


The New England Journal of Medicine | 2014

A Modified γ-Retrovirus Vector for X-Linked Severe Combined Immunodeficiency

Salima Hacein-Bey-Abina; Sung-Yun Pai; H. Bobby Gaspar; Myriam Armant; Charles C. Berry; Stéphane Blanche; Jack Bleesing; Johanna Blondeau; Helen de Boer; Karen Buckland; Laure Caccavelli; Guilhem Cros; Satir De Oliveira; Karen S. Fernández; Dongjing Guo; Chad E. Harris; Gregory Hopkins; Leslie Lehmann; Annick Lim; Wendy B. London; Johannes C.M. van der Loo; Nirav Malani; Frances Male; Punam Malik; M. Angélica Marinovic; Anne Marie McNicol; Despina Moshous; Bénédicte Neven; Matías Oleastro; Capucine Picard

BACKGROUND In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).


JAMA | 2015

Outcomes Following Gene Therapy in Patients With Severe Wiskott-Aldrich Syndrome

Salima Hacein-Bey Abina; H. Bobby Gaspar; Johanna Blondeau; Laure Caccavelli; Sabine Charrier; Karen Buckland; Capucine Picard; Emmanuelle Six; Nourredine Himoudi; Kimberly Gilmour; Anne-Marie McNicol; Havinder Hara; Jinhua Xu-Bayford; Christine Rivat; Fabien Touzot; Fulvio Mavilio; Annick Lim; Jean-Marc Tréluyer; Sébastien Héritier; François Lefrere; Isabelle Pengue-Koyi; Géraldine Honnet; Stéphane Blanche; Eric Sherman; Frances Male; Charles C. Berry; Nirav Malani; Frederic D. Bushman; Alain Fischer; Aj Thrasher

IMPORTANCE Wiskott-Aldrich syndrome is a rare primary immunodeficiency associated with severe microthrombocytopenia. Partially HLA antigen-matched allogeneic hematopoietic stem cell (HSC) transplantation is often curative but is associated with significant comorbidity. OBJECTIVE To assess the outcomes and safety of autologous HSC gene therapy in Wiskott-Aldrich syndrome. DESIGN, SETTING, AND PARTICIPANTS Gene-corrected autologous HSCs were infused in 7 consecutive patients with severe Wiskott-Aldrich syndrome lacking HLA antigen-matched related or unrelated HSC donors (age range, 0.8-15.5 years; mean, 7 years) following myeloablative conditioning. Patients were enrolled in France and England and treated between December 2010 and January 2014. Follow-up of patients in this intermediate analysis ranged from 9 to 42 months. INTERVENTION A single infusion of gene-modified CD34+ cells with an advanced lentiviral vector. MAIN OUTCOMES AND MEASURES Primary outcomes were improvement at 24 months in eczema, frequency and severity of infections, bleeding tendency, and autoimmunity and reduction in disease-related days of hospitalization. Secondary outcomes were improvement in immunological and hematological characteristics and evidence of safety through vector integration analysis. RESULTS Six of the 7 patients were alive at the time of last follow-up (mean and median follow-up, 28 months and 27 months, respectively) and showed sustained clinical benefit. One patient died 7 months after treatment of preexisting drug-resistant herpes virus infection. Eczema and susceptibility to infections resolved in all 6 patients. Autoimmunity improved in 5 of 5 patients. No severe bleeding episodes were recorded after treatment, and at last follow-up, all 6 surviving patients were free of blood product support and thrombopoietic agonists. Hospitalization days were reduced from a median of 25 days during the 2 years before treatment to a median of 0 days during the 2 years after treatment. All 6 surviving patients exhibited high-level, stable engraftment of functionally corrected lymphoid cells. The degree of myeloid cell engraftment and of platelet reconstitution correlated with the dose of gene-corrected cells administered. No evidence of vector-related toxicity was observed clinically or by molecular analysis. CONCLUSIONS AND RELEVANCE This study demonstrated the feasibility of the use of gene therapy in patients with Wiskott-Aldrich syndrome. Controlled trials with larger numbers of patients are necessary to assess long-term outcomes and safety.


Journal of Clinical Investigation | 2014

A human immunodeficiency caused by mutations in the PIK3R1 gene

Marie-Céline Deau; Lucie Heurtier; Pierre Frange; Felipe Suarez; Christine Bole-Feysot; Patrick Nitschke; Marina Cavazzana; Capucine Picard; Anne Durandy; Alain Fischer; Sven Kracker

Recently, patient mutations that activate PI3K signaling have been linked to a primary antibody deficiency. Here, we used whole-exome sequencing and characterized the molecular defects in 4 patients from 3 unrelated families diagnosed with hypogammaglobulinemia and recurrent infections. We identified 2 different heterozygous splice site mutations that affect the same splice site in PIK3R1, which encodes the p85α subunit of PI3K. The resulting deletion of exon 10 produced a shortened p85α protein that lacks part of the PI3K p110-binding domain. The hypothetical loss of p85α-mediated inhibition of p110 activity was supported by elevated phosphorylation of the known downstream signaling kinase AKT in patient T cell blasts. Analysis of patient blood revealed that naive T and memory B cell counts were low, and T cell blasts displayed enhanced activation-induced cell death, which was corrected by addition of the PI3Kδ inhibitor IC87114. Furthermore, B lymphocytes proliferated weakly in response to activation via the B cell receptor and TLR9, indicating a B cell defect. The phenotype exhibited by patients carrying the PIK3R1 splice site mutation is similar to that of patients carrying gain-of-function mutations in PIK3CD. Our results suggest that PI3K activity is tightly regulated in T and B lymphocytes and that various defects in the PI3K-triggered pathway can cause primary immunodeficiencies.


Haematologica | 2014

Hematopoietic stem cell transplantation in thalassemia major and sickle cell disease: indications and management recommendations from an international expert panel

Emanuele Angelucci; Susanne Matthes-Martin; Donatella Baronciani; Françoise Bernaudin; Sonia Bonanomi; Maria Domenica Cappellini; Jean Hugues Dalle; Paolo Di Bartolomeo; Cristina Díaz de Heredia; Roswitha Dickerhoff; Claudio Giardini; Eliane Gluckman; Ayad Achmed Hussein; Naynesh Kamani; Milen Minkov; Franco Locatelli; Vanderson Rocha; Petr Sedlacek; Frans Smiers; Isabelle Thuret; Isaac Yaniv; Marina Cavazzana; Christina Peters

Thalassemia major and sickle cell disease are the two most widely disseminated hereditary hemoglobinopathies in the world. The outlook for affected individuals has improved in recent years due to advances in medical management in the prevention and treatment of complications. However, hematopoietic stem cell transplantation is still the only available curative option. The use of hematopoietic stem cell transplantation has been increasing, and outcomes today have substantially improved compared with the past three decades. Current experience world-wide is that more than 90% of patients now survive hematopoietic stem cell transplantation and disease-free survival is around 80%. However, only a few controlled trials have been reported, and decisions on patient selection for hematopoietic stem cell transplantation and transplant management remain principally dependent on data from retrospective analyses and on the clinical experience of the transplant centers. This consensus document from the European Blood and Marrow Transplantation Inborn Error Working Party and the Paediatric Diseases Working Party aims to report new data and provide consensus-based recommendations on indications for hematopoietic stem cell transplantation and transplant management.


The New England Journal of Medicine | 2017

Gene Therapy in a Patient with Sickle Cell Disease.

Jean-Antoine Ribeil; Salima Hacein-Bey-Abina; Emmanuel Payen; Alessandra Magnani; Michaela Semeraro; Elisa Magrin; Laure Caccavelli; Bénédicte Neven; Philippe Bourget; Wassim El Nemer; Pablo Bartolucci; Leslie Weber; Hervé Puy; Jean-François Meritet; David Grevent; Yves Beuzard; Stany Chrétien; Thibaud Lefebvre; Robert W. Ross; Olivier Negre; Gabor Veres; Laura Sandler; Sandeep Soni; Mariane de Montalembert; Stéphane Blanche; Philippe Leboulch; Marina Cavazzana

Abstract Sickle cell disease results from a homozygous missense mutation in the β‐globin gene that causes polymerization of hemoglobin S. Gene therapy for patients with this disorder is complicated by the complex cellular abnormalities and challenges in achieving effective, persistent inhibition of polymerization of hemoglobin S. We describe our first patient treated with lentiviral vector–mediated addition of an antisickling β‐globin gene into autologous hematopoietic stem cells. Adverse events were consistent with busulfan conditioning. Fifteen months after treatment, the level of therapeutic antisickling β‐globin remained high (approximately 50% of β‐like–globin chains) without recurrence of sickle crises and with correction of the biologic hallmarks of the disease. (Funded by Bluebird Bio and others; HGB‐205 ClinicalTrials.gov number, NCT02151526.)


Blood | 2017

Sickle cell disease: An international survey of results of HLA-identical sibling hematopoietic stem cell transplantation

Eliane Gluckman; Barbara Cappelli; Françoise Bernaudin; Myriam Labopin; Fernanda Volt; Jeanette Carreras; Belinda Pinto Simões; Alina Ferster; Sophie Dupont; Josu de la Fuente; Jean-Hugues Dalle; Marco Zecca; Mark C. Walters; Lakshmanan Krishnamurti; Monica Bhatia; Kathryn Leung; Gregory Yanik; Joanne Kurtzberg; Nathalie Dhedin; Mathieu Kuentz; Gérard Michel; Jane F. Apperley; Patrick Lutz; Bénédicte Neven; Yves Bertrand; Jean Pierre Vannier; Mouhab Ayas; Marina Cavazzana; Susanne Matthes-Martin; Vanderson Rocha

Despite advances in supportive therapy to prevent complications of sickle cell disease (SCD), access to care is not universal. Hematopoietic cell transplantation is, to date, the only curative therapy for SCD, but its application is limited by availability of a suitable HLA-matched donor and lack of awareness of the benefits of transplant. Included in this study are 1000 recipients of HLA-identical sibling transplants performed between 1986 and 2013 and reported to the European Society for Blood and Marrow Transplantation, Eurocord, and the Center for International Blood and Marrow Transplant Research. The primary endpoint was event-free survival, defined as being alive without graft failure; risk factors were studied using a Cox regression models. The median age at transplantation was 9 years, and the median follow-up was longer than 5 years. Most patients received a myeloablative conditioning regimen (n = 873; 87%); the remainder received reduced-intensity conditioning regimens (n = 125; 13%). Bone marrow was the predominant stem cell source (n = 839; 84%); peripheral blood and cord blood progenitors were used in 73 (7%) and 88 (9%) patients, respectively. The 5-year event-free survival and overall survival were 91.4% (95% confidence interval, 89.6%-93.3%) and 92.9% (95% confidence interval, 91.1%-94.6%), respectively. Event-free survival was lower with increasing age at transplantation (hazard ratio [HR], 1.09; P < .001) and higher for transplantations performed after 2006 (HR, 0.95; P = .013). Twenty-three patients experienced graft failure, and 70 patients (7%) died, with the most common cause of death being infection. The excellent outcome of a cohort transplanted over the course of 3 decades confirms the role of HLA-identical sibling transplantation for children and adults with SCD.


Blood | 2014

SCID patients with ARTEMIS vs RAG deficiencies following HCT: increased risk of late toxicity in ARTEMIS-deficient SCID.

Catharina Schuetz; Bénédicte Neven; Christopher C. Dvorak; Sandrine Leroy; Markus Ege; Ulrich Pannicke; Klaus Schwarz; Ansgar Schulz; Manfred Hoenig; Monika Sparber-Sauer; Susanne A. Gatz; Christian Denzer; Stéphane Blanche; Despina Moshous; Capucine Picard; Biljana Horn; Jean-Pierre de Villartay; Marina Cavazzana; Klaus-Michael Debatin; Wilhelm Friedrich; Alain Fischer; Morton J. Cowan

A subgroup of severe combined immunodeficiencies (SCID) is characterized by lack of T and B cells and is caused by defects in genes required for T- and B-cell receptor gene rearrangement. Several of these genes are also involved in nonhomologous end joining of DNA double-strand break repair, the largest subgroup consisting of patients with T(-)B(-)NK(+)SCID due to DCLRE1C/ARTEMIS defects. We postulated that in patients with ARTEMIS deficiency, early and late complications following hematopoietic cell transplantation might be more prominent compared with patients with T(-)B(-)NK(+)SCID caused by recombination activating gene 1/2 (RAG1/2) deficiencies. We analyzed 69 patients with ARTEMIS and 76 patients with RAG1/2 deficiencies who received transplants from either HLA-identical donors without conditioning or from HLA-nonidentical donors without or with conditioning. There was no difference in survival or in the incidence or severity of acute graft-versus-host disease regardless of exposure to alkylating agents. Secondary malignancies were not observed. Immune reconstitution was comparable in both groups, however, ARTEMIS-deficient patients had a significantly higher occurrence of infections in long-term follow-up. There is a highly significant association between poor growth in ARTEMIS deficiency and use of alkylating agents. Furthermore, abnormalities in dental development and endocrine late effects were associated with alkylation therapy in ARTEMIS deficiency.


Human Gene Therapy | 2016

Gene Therapy of the β-Hemoglobinopathies by Lentiviral Transfer of the βA(T87Q)-Globin Gene

Olivier Negre; Anne-Virginie Eggimann; Yves Beuzard; Jean-Antoine Ribeil; Philippe Bourget; Suparerk Borwornpinyo; Suradej Hongeng; Salima Hacein-Bey; Marina Cavazzana; Philippe Leboulch; Emmanuel Payen

β-globin gene disorders are the most prevalent inherited diseases worldwide and result from abnormal β-globin synthesis or structure. Novel therapeutic approaches are being developed in an effort to move beyond palliative management. Gene therapy, by ex vivo lentiviral transfer of a therapeutic β-globin gene derivative (βAT87Q-globin) to hematopoietic stem cells, driven by cis-regulatory elements that confer high, erythroid-specific expression, has been evaluated in human clinical trials over the past 8 years. βAT87Q-globin is used both as a strong inhibitor of HbS polymerization and as a biomarker. While long-term studies are underway in multiple centers in Europe and in the United States, proof-of-principle of efficacy and safety has already been obtained in multiple patients with β-thalassemia and sickle cell disease.


Expert Opinion on Biological Therapy | 2014

Gene therapy for inherited immunodeficiency

Fabien Touzot; Salima Hacein-Bey-Abina; Alain Fischer; Marina Cavazzana

Introduction: During the last decade, gene therapy has emerged as a convincing therapy for primary immunodeficiencies (PIDs). Ex vivo gene transfer into autologous hematopoietic stem cells (HSCs) via viral vectors permits sustained correction of T cell immunodeficiency in two forms of severe combined immunodeficiency: X–linked SCID (SCID-X1) (γ chain [γc] deficiency) and adenosine deaminase deficiency. However, this success has been balanced by the occurrence of genotoxicity generated by the integration of first-generation retroviral vectors. Recently, the development of safer self-inactivating vectors has led to the initiation of new studies with the hope of equivalent efficacy and a better safety profile. Areas covered: This review article focuses on the updated results of gene therapy trials for PIDs – from early studies to ongoing clinical trials. We detail the major advances made in gene transfer and repair technologies, and discuss the many ways to extend our present experience. Expert opinion: With optimization in terms of safety and efficacy, gene therapy by lentiviral transduction could become a compelling alternative to allogeneic HSC transplantation, and thus may take center stage in the management of PIDs in coming years.


Blood | 2015

Faster T-cell development following gene therapy compared with haploidentical HSCT in the treatment of SCID-X1

Fabien Touzot; Despina Moshous; Rita Creidy; Bénédicte Neven; Pierre Frange; Guilhem Cros; Laure Caccavelli; Johanna Blondeau; Alessandra Magnani; Jean-Marc Luby; Brigitte Ternaux; Capucine Picard; Stéphane Blanche; Alain Fischer; Salima Hacein-Bey-Abina; Marina Cavazzana

During the last decade, gene therapy via ex vivo gene transfer into autologous hematopoietic stem cells has emerged as a convincing therapy for severe combined immunodeficiency caused by ILR2G mutation (SCID-X1) despite the occurrence of genotoxicity caused by the integration of first-generation retroviral vectors. However, the place of gene therapy among the therapeutic armamentarium remains to be defined. We retrospectively analyze and compare clinical outcomes and immune reconstitution in 13 consecutive SCID-X1 patients having undergone haploidentical hematopoietic stem cell transplantation (HSCT) and 14 SCID-X1 patients treated with gene therapy over the same period at a single center level: the Necker Childrens Hospital (Paris, France). Our results show a clear advantage in terms of T-cell development of gene therapy over HSCT with a mismatched donor. Patients treated with gene therapy display a faster T-cell reconstitution and a better long-term thymic output. Interestingly, this advantage of gene therapy vs haploidentical HSCT seems to be independent of the existence of clinical graft-versus-host disease in the latter condition. If data of safety are confirmed over the long term, gene therapy for SCID-X1 appears to be an equal, if not superior, alternative to haploidentical HSCT.

Collaboration


Dive into the Marina Cavazzana's collaboration.

Top Co-Authors

Avatar

Alain Fischer

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Despina Moshous

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Stéphane Blanche

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Bénédicte Neven

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar

Fabien Touzot

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philippe Leboulch

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Capucine Picard

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Jean-Antoine Ribeil

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Felipe Suarez

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge