Network


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

Hotspot


Dive into the research topics where Marina Cavazzana-Calvo is active.

Publication


Featured researches published by Marina Cavazzana-Calvo.


Nature | 2010

Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia

Marina Cavazzana-Calvo; Emmanuel Payen; Olivier Negre; Gary P. Wang; Kathleen Hehir; Floriane Fusil; Julian D. Down; Maria Denaro; Troy Brady; Karen A. Westerman; Resy Cavallesco; Beatrix Gillet-Legrand; Laure Caccavelli; Riccardo Sgarra; Leila Maouche-Chretien; Françoise Bernaudin; Robert Girot; Ronald Dorazio; Geert Jan Mulder; Axel Polack; Arthur Bank; Jean Soulier; Jérôme Larghero; Nabil Kabbara; Bruno Dalle; Bernard Gourmel; Gérard Socié; Stany Chrétien; Nathalie Cartier; Patrick Aubourg

The β-haemoglobinopathies are the most prevalent inherited disorders worldwide. Gene therapy of β-thalassaemia is particularly challenging given the requirement for massive haemoglobin production in a lineage-specific manner and the lack of selective advantage for corrected haematopoietic stem cells. Compound βE/β0-thalassaemia is the most common form of severe thalassaemia in southeast Asian countries and their diasporas. The βE-globin allele bears a point mutation that causes alternative splicing. The abnormally spliced form is non-coding, whereas the correctly spliced messenger RNA expresses a mutated βE-globin with partial instability. When this is compounded with a non-functional β0 allele, a profound decrease in β-globin synthesis results, and approximately half of βE/β0-thalassaemia patients are transfusion-dependent. The only available curative therapy is allogeneic haematopoietic stem cell transplantation, although most patients do not have a human-leukocyte-antigen-matched, geno-identical donor, and those who do still risk rejection or graft-versus-host disease. Here we show that, 33 months after lentiviral β-globin gene transfer, an adult patient with severe βE/β0-thalassaemia dependent on monthly transfusions since early childhood has become transfusion independent for the past 21 months. Blood haemoglobin is maintained between 9 and 10 g dl−1, of which one-third contains vector-encoded β-globin. Most of the therapeutic benefit results from a dominant, myeloid-biased cell clone, in which the integrated vector causes transcriptional activation of HMGA2 in erythroid cells with further increased expression of a truncated HMGA2 mRNA insensitive to degradation by let-7 microRNAs. The clonal dominance that accompanies therapeutic efficacy may be coincidental and stochastic or result from a hitherto benign cell expansion caused by dysregulation of the HMGA2 gene in stem/progenitor cells.


Cell | 2001

Artemis, a novel DNA double-strand break repair/V(D)J recombination protein, is mutated in human severe combined immune deficiency.

Despina Moshous; Isabelle Callebaut; Régina de Chasseval; Barbara Corneo; Marina Cavazzana-Calvo; Françoise Le Deist; Ilhan Tezcan; Ozden Sanal; Yves Bertrand; Noël Philippe; Alain Fischer; Jean-Pierre de Villartay

The V(D)J recombination process insures the somatic diversification of immunoglobulin and antigen T cell receptor encoding genes. This reaction is initiated by a DNA double-strand break (dsb), which is resolved by the ubiquitously expressed DNA repair machinery. Human T-B-severe combined immunodeficiency associated with increased cellular radiosensitivity (RS-SCID) is characterized by a defect in the V(D)J recombination leading to an early arrest of both B and T cell maturation. We previously mapped the disease-related locus to the short arm of chromosome 10. We herein describe the cloning of the gene encoding a novel protein involved in V(D)J recombination/DNA repair, Artemis, whose mutations cause human RS-SCID. Protein sequence analysis strongly suggests that Artemis belongs to the metallo-beta-lactamase superfamily.


The Lancet | 2003

Long-term survival and transplantation of haemopoietic stem cells for immunodeficiencies: report of the European experience 1968–99

Corinne Antoine; Susanna M. Müller; Andrew J. Cant; Marina Cavazzana-Calvo; Paul Veys; Jaak M. Vossen; Anders Fasth; Carsten Heilmann; N Wulffraat; Reinhard Seger; Stéphane Blanche; Wilhelm Friedrich; Mario Abinun; Graham Davies; Robert Bredius; Ansgar Schulz; Paul Landais; Alain Fischer

BACKGROUND Transplantation of allogeneic haemopoietic stem cells can cure several primary immunodeficiencies. This European report focuses on the long-term results of such procedures done between 1968 and December, 1999, for primary immunodeficiencies. METHODS The report includes data from 37 centres in 18 countries, which participated in a European registry for stem-cell transplantation in severe combined immuno deficiencies (SCID) and in other immunodeficiency disorders (non-SCID). 1082 transplants in 919 patients were studied (566 in 475 SCID patients, 512 in 444 non-SCID patients; four procedures excluded owing to insufficient data). Minimum follow-up of 6 months was required. FINDINGS In SCID, 3-year survival with sustained engraftment was significantly better after HLA-identical than after mismatched transplantation (77% vs 54%; p=0.002) and survival improved over time. In HLA-mismatched stem-cell transplantation, B(-) SCID had poorer prognosis than B(+) SCID. However, improvement with time occurred in both SCID phenotypes. In non-SCID, 3-year survival after genotypically HLA-matched, phenotypically HLA-matched, HLA-mismatched related, and unrelated-donor transplantation was 71%, 42%, 42%, and 59%, respectively (p=0.0006). Acute graft versus host disease predicted poor prognosis whatever the donor origin except in related HLA-identical transplantation in SCID. INTERPRETATION The improvement in survival over time indicates more effective prevention and treatment of disease-related and procedure-related complications--eg, infections and graft versus host disease. An important factor is better prevention of graft versus host disease in the HLA-non-identical setting by use of more efficient methods of T-cell depletion. For non-SCID, stem-cell transplantation can provide a cure, and grafts from unrelated donors are almost as beneficial as those from genetically HLA-identical relatives.


The Journal of Allergy and Clinical Immunology | 2010

Transplantation of hematopoietic stem cells and long-term survival for primary immunodeficiencies in Europe: Entering a new century, do we do better?

Andrew R. Gennery; Mary Slatter; Laure Grandin; Pierre Taupin; Andrew J. Cant; Paul Veys; Persis Amrolia; H. Bobby Gaspar; E. Graham Davies; Wilhelm Friedrich; Manfred Hoenig; Luigi D. Notarangelo; Evelina Mazzolari; Fulvio Porta; Robbert G. M. Bredius; Arjen C. Lankester; Nico Wulffraat; Reinhard Seger; Tayfun Güngör; Anders Fasth; Petr Sedlacek; Bénédicte Neven; Stéphane Blanche; Alain Fischer; Marina Cavazzana-Calvo; Paul Landais

BACKGROUND Hematopoietic stem cell transplantation remains the only treatment for most patients with severe combined immunodeficiencies (SCIDs) or other primary immunodeficiencies (non-SCID PIDs). OBJECTIVE To analyze the long-term outcome of patients with SCID and non-SCID PID from European centers treated between 1968 and 2005. METHODS The product-limit method estimated cumulative survival; the log-rank test compared survival between groups. A Cox proportional-hazard model evaluated the impact of independent predictors on patient survival. RESULTS In patients with SCID, survival with genoidentical donors (n = 25) from 2000 to 2005 was 90%. Survival using a mismatched relative (n = 96) has improved (66%), similar to that using an unrelated donor (n = 46; 69%; P = .005). Transplantation after year 1995, a younger age, B(+) phenotype, genoidentical and phenoidentical donors, absence of respiratory impairment, or viral infection before transplantation were associated with better prognosis on multivariate analysis. For non-SCID PID, in contrast with patients with SCID, we confirm that, in the 2000 to 2005 period, using an unrelated donor (n = 124) gave a 3-year survival rate similar to a genoidentical donor (n = 73), 79% for both. Survival was 76% in phenoidentical transplants (n = 23) and worse in mismatched related donor transplants (n = 47; 46%; P = .016). CONCLUSION This is the largest cohort study of such patients with the longest follow-up. Specific issues arise for different patient groups. Patients with B-SCID have worse survival than other patients with SCID, despite improvements in each group. For non-SCID PID, survival is worse than SCID, although more conditions are now treated. Individual disease categories now need to be analyzed so that disease-specific prognosis may be better understood and the best treatments planned.


Journal of Clinical Investigation | 2007

Vector integration is nonrandom and clustered and influences the fate of lymphopoiesis in SCID-X1 gene therapy

Annette Deichmann; Salima Hacein-Bey-Abina; Manfred Schmidt; Alexandrine Garrigue; Martijn H. Brugman; Jingqiong Hu; Hanno Glimm; Gabor Gyapay; Bernard Prum; Christopher C. Fraser; Nicolas Fischer; Kerstin Schwarzwaelder; Maria Luise Siegler; Dick de Ridder; Karin Pike-Overzet; Steven J. Howe; Adrian J. Thrasher; Gerard Wagemaker; Ulrich Abel; Frank J. T. Staal; Eric Delabesse; Jean Luc Villeval; Bruce J. Aronow; Christophe Hue; Claudia Prinz; Manuela Wissler; Chuck Klanke; Jean Weissenbach; Ian E. Alexander; Alain Fischer

Recent reports have challenged the notion that retroviruses and retroviral vectors integrate randomly into the host genome. These reports pointed to a strong bias toward integration in and near gene coding regions and, for gammaretroviral vectors, around transcription start sites. Here, we report the results obtained from a large-scale mapping of 572 retroviral integration sites (RISs) isolated from cells of 9 patients with X-linked SCID (SCID-X1) treated with a retrovirus-based gene therapy protocol. Our data showed that two-thirds of insertions occurred in or very near to genes, of which more than half were highly expressed in CD34(+) progenitor cells. Strikingly, one-fourth of all integrations were clustered as common integration sites (CISs). The highly significant incidence of CISs in circulating T cells and the nature of their locations indicate that insertion in many gene loci has an influence on cell engraftment, survival, and proliferation. Beyond the observed cases of insertional mutagenesis in 3 patients, these data help to elucidate the relationship between vector insertion and long-term in vivo selection of transduced cells in human patients with SCID-X1.


The Lancet | 2002

Immune reconstitution without graft-versus-host disease after haemopoietic stem-cell transplantation: a phase 1/2 study

Isabelle André-Schmutz; Françoise Le Deist; Salima Hacein-Bey-Abina; Ellen S. Vitetta; Gaëlle Chédeville; Etienne Vilmer; Alain Fischer; Marina Cavazzana-Calvo

BACKGROUND Allogeneic haemopoietic stem-cell transplantation (HSCT) is the treatment of choice for many haematological malignancies and inherited disorders. When stem cells for transplantation come from a human leucocyte antigen matched unrelated donor, or from a partly mismatched related donor, ex-vivo T-cell depletion of the graft can prevent development of graft-versus-host disease, but lead in turn to a delay in immune reconstitution and a concordant increase in incidence of opportunistic infections and leukaemic relapses. We aimed to infuse T cells selectively depleted in allogeneic T cells that cause graft-versus-host disease using an ex-vivo procedure designed to eliminate alloactivated donor T cells, with an immunotoxin that reacts with a cell surface activation antigen, CD25. METHODS We did a phase 1/2 study, in which 1-8 x 10(5) allodepleted T cells/kg were infused between days 15 and 47 into 15 paediatric patients who had acquired or congenital haemopoietic disorders and who received HSCT on day 0. Occurrence of graft-versus-host disease and time to immune reconstitution were assessed. No treatment for graft-versus-host disease was given. FINDINGS Less than 1% residual anti-host alloreactivity was recorded in 12 of 16 procedures. Other immune responses were preserved by the allodepletion procedure in 12 cases. No cases of severe (greater than grade II) graft-versus-host disease arose. Evidence for early T-cell expansion was shown in three patients with continuing viral infections. Specific antiviral responses, such as strong cytolytic activity, were noted. INTERPRETATION Our results show that ex-vivo selective depletion of T cells that cause graft-versus-host disease is efficient and feasible, even in haploidentical settings.


Nature | 2004

The future of gene therapy.

Marina Cavazzana-Calvo; Adrian J. Thrasher; Fulvio Mavilio

Balancing the risks and benefits of clinical trial.


Immunological Reviews | 2005

Severe combined immunodeficiency. A model disease for molecular immunology and therapy

Alain Fischer; Françoise Le Deist; Salima Hacein-Bey-Abina; Isabelle André-Schmutz; Geneviève de Saint Basile; Jean-Pierre de Villartay; Marina Cavazzana-Calvo

Summary:  Severe combined immunodeficiencies (SCIDs) consist of genetically determined arrest of T‐cell differentiation. Ten different molecular defects have now been identified, which all lead to early death in the absence of therapy. Transplantation of allogeneic hematopoietic stem cells (HSCT) can restore T‐cell development, thus saving the lives of SCID patients. In this review, the different characteristics of HSCT are discussed along with the available data regarding the long‐term outcome. Transient thymopoiesis caused by an exhaustion of donor progenitor cells and possibly a progressive loss of thymus function can lead to a progressive decline in T‐cell functions. The preliminary results of gene therapy show the correction of two SCID conditions. Based on the assumption that long‐lasting pluripotent progenitor cells are transduced, these data suggest that gene therapy could overcome the long‐term recurrence of the T‐cell immunodeficiency. SCID is thus a disease model for experimental therapy in the hematopoietic system.


Journal of Clinical Investigation | 2007

Gene therapy for severe combined immunodeficiency: are we there yet?

Marina Cavazzana-Calvo; Alain Fischer

Inherited and acquired diseases of the hematopoietic system can be cured by allogeneic hematopoietic stem cell transplantation. This treatment strategy is highly successful when an HLA-matched sibling donor is available, but if not, few therapeutic options exist. Gene-modified, autologous bone marrow transplantation can circumvent the severe immunological complications that occur when a related HLA-mismatched donor is used and thus represents an attractive alternative. In this review, we summarize the advantages and limitations associated with the use of gene therapy to cure SCID. Insertional mutagenesis and technological improvements aimed at increasing the safety of this strategy are also discussed.


Cell Stem Cell | 2008

New ISSCR Guidelines Underscore Major Principles for Responsible Translational Stem Cell Research

Insoo Hyun; Olle Lindvall; Lars Ährlund-Richter; Marina Cavazzana-Calvo; Giulio Cossu; Michele De Luca; Ira J. Fox; Claude Gerstle; Robert A. Goldstein; Göran Hermerén; Katherine A. High; Hyun Ok Kim; Hin-Peng Lee; Ephrat Levy-Lahad; Lingsong Li; Bernard Lo; Daniel Robert Marshak; Angela McNab; Megan Munsie; Hiromitsu Nakauchi; Mahendra S. Rao; Heather M. Rooke; Carlos Simon Valles; Alok Srivastava; Jeremy Sugarman; Patrick L. Taylor; Anna Veiga; Adrianne L. Wong; Laurie Zoloth; George Q. Daley

The International Society for Stem Cell Research (ISSCR) task force that developed new Guidelines for the Clinical Translation of Stem Cells discusses core principles that should guide the responsible transition of basic stem cell research into appropriate clinical applications.

Collaboration


Dive into the Marina Cavazzana-Calvo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stéphane Blanche

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Isabelle André-Schmutz

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Salima Hacein-Bey

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

F. Le Deist

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Paul Veys

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Capucine Picard

Paris Descartes University

View shared research outputs
Researchain Logo
Decentralizing Knowledge