Network


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

Hotspot


Dive into the research topics where Giovanna Lucchini is active.

Publication


Featured researches published by Giovanna Lucchini.


Science Translational Medicine | 2017

Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells

Waseem Qasim; Hong Zhan; Sujith Samarasinghe; Stuart Adams; Persis Amrolia; Sian Stafford; Katie Butler; C Rivat; Gary Wright; K Somana; Sara Ghorashian; Danielle Pinner; Gul Ahsan; Kimberly Gilmour; Giovanna Lucchini; S Inglott; W Mifsud; Robert Chiesa; Karl S. Peggs; L Chan; F Farzeneh; Adrian J. Thrasher; Ajay Vora; Martin Pule; Paul Veys

Universal gene-edited CAR19 T cells eliminate infant leukemia. CAR sharing Chimeric antigen receptor (CAR) T cells can be very effective in treating acute lymphocytic leukemia. Unfortunately, these therapeutic cells have to be custom-made for each patient, and this is not always feasible, especially for patients who do not have sufficient healthy T cells. Qasim et al. demonstrate that there may be another option for these patients. By using gene editing to simultaneously introduce the CAR and disrupt TCR and CD52 in T cells, the authors generated functional CAR T cells that could evade host immunity for use in unmatched recipients. These “off-the-shelf” CAR T cells were then used to treat two infants with relapsed refractory acute lymphocytic leukemia and bridge them to allogeneic stem cell transplantation. Autologous T cells engineered to express chimeric antigen receptor against the B cell antigen CD19 (CAR19) are achieving marked leukemic remissions in early-phase trials but can be difficult to manufacture, especially in infants or heavily treated patients. We generated universal CAR19 (UCART19) T cells by lentiviral transduction of non–human leukocyte antigen–matched donor cells and simultaneous transcription activator-like effector nuclease (TALEN)–mediated gene editing of T cell receptor α chain and CD52 gene loci. Two infants with relapsed refractory CD19+ B cell acute lymphoblastic leukemia received lymphodepleting chemotherapy and anti-CD52 serotherapy, followed by a single-dose infusion of UCART19 cells. Molecular remissions were achieved within 28 days in both infants, and UCART19 cells persisted until conditioning ahead of successful allogeneic stem cell transplantation. This bridge-to-transplantation strategy demonstrates the therapeutic potential of gene-editing technology.


Bone Marrow Transplantation | 2013

Feasibility of IL-15-activated cytokine-induced killer cell infusions after haploidentical stem cell transplantation

Eva Rettinger; Halvard Bonig; S Wehner; Giovanna Lucchini; Andre Willasch; Andrea Jarisch; Jan Soerensen; R Esser; Claudia Rossig; Thomas Klingebiel; Peter Bader

Feasibility of IL-15-activated cytokine-induced killer cell infusions after haploidentical stem cell transplantation


Haematologica | 2016

Mesenchymal stromal cells generated from pooled mononuclear cells of multiple bone marrow donors as a rescue therapy for children with severe steroid-refractory graft versus host disease: a multicenter survey

Zyrafete Kuçi; Halvard Bonig; Hermann Kreyenberg; Milica Bunos; Anna Jauch; Johannes W.G. Janssen; Marijana Skific; Kristina Michel; Ben Eising; Giovanna Lucchini; Shahrzad Bakhtiar; Johann Greil; Peter J. Lang; Oliver Basu; Irene von Luettichau; Ansgar Schultz; Karl-Walter Sykora; Andrea Jarisch; Jan Soerensen; Emilia Salzmann-Manrique; Erhard Seifried; Thomas Klingebiel; Peter Bader; Selim Kuçi

To circumvent donor-to-donor heterogeneity which may lead to inconsistent results after treatment of acute graft-versus-host disease with mesenchymal stromal cells generated from single donors we developed a novel approach by generating these cells from pooled bone marrow mononuclear cells of 8 healthy “3rd-party” donors. Generated cells were frozen in 209 vials and designated as mesenchymal stromal cell bank. These vials served as a source for generation of clinical grade mesenchymal stromal cell end-products, which exhibited typical mesenchymal stromal cell phenotype, trilineage differentiation potential and at later passages expressed replicative senescence-related markers (p21 and p16). Genetic analysis demonstrated their genomic stability (normal karyotype and a diploid pattern). Importantly, clinical end-products exerted a significantly higher allosuppressive potential than the mean allosuppressive potential of mesenchymal stromal cells generated from the same donors individually. Administration of 81 mesenchymal stromal cell end-products to 26 patients with severe steroid-resistant acute graft-versus-host disease in 7 stem cell transplant centers who were refractory to many lines of treatment, induced a 77% overall response at the primary end point (day 28). Remarkably, although the cohort of patients was highly challenging (96% grade III/IV and only 4% grade II graft-versus-host disease), after treatment with mesenchymal stromal cell end-products the overall survival rate at two years follow up was 71±11% for the entire patient cohort, compared to 51.4±9.0% in graft-versus-host disease clinical studies, in which mesenchymal stromal cells were derived from single donors. Mesenchymal stromal cell end-products may, therefore, provide a novel therapeutic tool for the effective treatment of severe acute graft-versus-host disease.


Cytotherapy | 2015

Immune reconstitution after cord blood transplantation: peculiarities, clinical implications and management strategies.

Giovanna Lucchini; Miguel-Angel Perales; Paul Veys

Umbilical cord blood (UCB) is now widely used as an alternative hematopoietic stem cell source for patients lacking closely matched related or unrelated adult donors. UCB transplantation has traditionally been associated with delayed engraftment, poor immune reconstitution and consequent increased risk of infection. More recent clinical studies, however, suggest that conditioning regimens and in particular the omission of in vivo T-cell depletion may play a crucial role in post-transplant T-cell expansion, facilitating a uniquely rapid immune recovery after UCB transplantation. The peculiar characteristics of UCB cells, the importance of thymic function and the role of conditioning regimens and graft-versus-host disease influencing immune reconstitution are described. The last part of the review reports available data on UCB, as well as third-party peripheral blood derived anti-viral cell therapy, which provides a novel approach to rescue UCB recipients with viral complications in the post-transplant period.


Blood | 2016

Stem cell transplantation for tetratricopeptide repeat domain 7A deficiency: long-term follow-up.

Jochen Kammermeier; Giovanna Lucchini; Sung-Yun Pai; Austen Worth; D Rampling; Persis Amrolia; Juliana Silva; Robert Chiesa; Kanchan Rao; Gabriele Noble-Jamieson; Marco Gasparetto; Drew Ellershaw; Holm H. Uhlig; Nj Sebire; Mamoun Elawad; Luigi D. Notarangelo; Neil P. Shah; Paul Veys

To the editor: Mutations in the tetratricopeptide repeat domain 7A ( TTC7A) gene cause a severe form of very early onset inflammatory bowel disease (VEOIBD).[1][1] TTC7A has a crucial role in chaperoning the enzyme phosphatidylinositol-4-kinase-3-α from the trans-Golgi apparatus to the plasma


Bone Marrow Transplantation | 2018

Effective treatment of steroid and therapy-refractory acute graft-versus-host disease with a novel mesenchymal stromal cell product (MSC-FFM)

Peter Bader; Zyrafete Kuçi; Shahrzad Bakhtiar; Oliver Basu; Gesine Bug; Michael Dennis; Johann Greil; Aniko Barta; Krisztián M. Kállay; Peter Lang; Giovanna Lucchini; Raj Pol; Ansgar Schulz; Karl-Walter Sykora; Irene von Luettichau; Grit Herter-Sprie; Mohammad Ashab Uddin; Phil Jenkin; Abdulrahman Alsultan; Jochen Buechner; Jerry Stein; Agnes Kelemen; Andrea Jarisch; Jan Soerensen; Emilia Salzmann-Manrique; Martin Hutter; Richard Schäfer; Erhard Seifried; Thomas Klingebiel; Halvard Bonig

The inability to generate mesenchymal stromal cells (MSCs) of consistent potency likely is responsible for inconsistent clinical outcomes of patients with aGvHD receiving MSC products. We developed a novel MSC manufacturing protocol characterized by high in vitro potency and near-identity of individual doses, referred to as “MSC-Frankfurt am Main (MSC-FFM)”. Herein, we report outcomes of the 69 patients who have received MSC-FFM. These were 51 children and 18 adults with refractory aGvHD grade II (4%), III (36%) or IV (59%). Patients were refractory either to frontline therapy (steroids) (29%) or to steroids and 1–5 additional lines of immunosuppressants (71%) were given infusions in four weekly intervals. The day 28 overall response rate was 83%; at the last follow-up, 61% and 25% of patients were in complete or partial remission. The median follow-up was 8.1 months. Six-month estimate for cumulative incidence of non-relapse mortality was 27% (range, 16–38); leukemia relapse mortality was 2% (range, 0–5). This was associated with a superior six-month overall survival (OS) probability rate of 71% (range, 61–83), compared to the outcome of patients not treated with MSC-FFM. This novel product was effective in children and adults, suggesting that MSC-FFM represents a promising therapy for steroid refractory aGvHD.


Biology of Blood and Marrow Transplantation | 2017

Impact of Conditioning Regimen on Outcomes for Children with Acute Myeloid Leukemia Undergoing Transplantation in First Complete Remission. An Analysis on Behalf of the Pediatric Disease Working Party of the European Group for Blood and Marrow Transplantation

Giovanna Lucchini; Myriam Labopin; Eric Beohou; Arnauld Dalissier; Jean Hughes Dalle; Jacqueline Cornish; Marco Zecca; Sujith Samarasinghe; Brenda Gibson; Franco Locatelli; Yves Bertrand; Fawzi Abdel-Rahman; Gerald Socie; Mikael Sundin; Arjan C. Lankester; Peter Sedlacek; Rose Marie Hamladji; Carsten Heilmann; Boris Afanasyev; Rachel Hough; C. Peters; Peter Bader; Paul Veys

Hematopoietic stem cell transplantation (HSCT) represents the cornerstone of treatment in pediatric high-risk and relapsed acute myeloid leukemia (AML). The aim of the present study was to compare outcomes of pediatric patients with AML undergoing HSCT using 3 different conditioning regimens: total body irradiation (TBI) and cyclophosphamide (Cy); busulfan (Bu) and Cy; or Bu, Cy, and melphalan (Mel). In this retrospective study, registry data for patients > 2 and <18 years age undergoing matched allogeneic HSCT for AML in first complete remission (CR1) in 204 European Group for Blood and Marrow Transplantation centers between 2000 and 2010 were analyzed. Data were available for 631 patients; 458 patients received stem cells from a matched sibling donor and 173 from a matched unrelated donor. For 440 patients, bone marrow was used as stem cell source, and 191 patients received peripheral blood stem cells. One hundred nine patients received TBICy, 389 received BuCy, and 133 received BuCyMel as their preparatory regimen. Median follow-up was 55 months. Patients receiving BuCyMel showed a lower incidence of relapse at 5 years (14.7% versus 31.5% in BuCy versus 30% in TBICy, P < .01) and higher overall survival (OS) (76.6% versus 64% versus 64.5%, P = .04) and leukemia-free survival (LFS) (74.5% versus 58% versus 61.9%, P < .01), with a comparable nonrelapse mortality (NRM) (10.8% versus 10.5% versus 8.1%, P = .79). Acute graft-versus-host disease (GVHD) grades III and IV but not chronic GVHD, was higher in patients receiving BuCyMel. Older age at HSCT had an adverse impact on NRM and the use of peripheral blood as stem cell source was associated with increased chronic GVHD and NRM as well as lower LFS and OS. Among pediatric patients receiving HSCT for AML in CR1, the use of BuCyMel conditioning proved superior to TBICy and BuCy in reducing relapse and improving LFS.


Current Opinion in Pediatrics | 2016

Evolving hematopoietic stem cell transplantation strategies in severe aplastic anemia.

Andrew C. Dietz; Giovanna Lucchini; Sujith Samarasinghe; Michael A. Pulsipher

Purpose of review Significant improvements in unrelated donor hematopoietic stem cell transplantation (HSCT) in recent years have solidified its therapeutic role in severe aplastic anemia (SAA) and led to the evolution of treatment algorithms, particularly for children. Recent findings Advances in understanding the genetics of inherited bone marrow failure syndromes (IBMFS) have allowed more confidence in accurately diagnosing SAA and avoiding treatments that could be dangerous and ineffective in individuals with IBMFS, which can be diagnosed in 10–20% of children presenting with a picture of SAA. Additionally long-term survival after matched sibling donor and matched unrelated donor HSCT now exceed 90% in children. Late effects after HSCT for SAA are minimal with current strategies, and compare favorably to late effects after upfront immunosuppressive therapy, except for patients with chronic graft versus host disease. Summary Careful assessment for signs or symptoms of IBMFS, along with genetic screening for these disorders, is of major importance. Matched sibling donor HSCT is already considered the standard of care for upfront therapy and some groups are evaluating matched unrelated donor HSCT as primary therapy. Ongoing studies will continue to challenge treatment algorithms and may lead to an even more expanded role for HSCT in SAA.


Expert Review of Hematology | 2014

Hematopoietic stem cell transplantation and immunotherapy for pediatric acute myeloid leukemia: an open challenge

Giovanna Lucchini; Peter Bader

The present review summarizes the role of immunotherapy in the treatment of pediatric acute myeloid leukemia (AML). Through the evaluation of recent clinical and pre-clinical studies, different approaches are taken into account: from the allogeneic transplantation as remission consolidation strategy, to the most advanced targeted cell therapy approaches for relapse prevention. The entangled question of immunotherapys evolving role in AML is also addressed, as new diagnostic tools provide better risk stratification, classic chemotherapy allows for higher survival rates and targeted drugs became available for children too. Moreover, the need for collaborative, prospective studies in this field is highlighted. Only through sound data it will be possible to compare and possibly integrate new therapies with conventional approaches, with the final aim of achieving a higher survival rate and a better quality of life for our patients.


Bone Marrow Transplantation | 2017

Allogeneic stem cell transplantation for refractory acute myeloid leukemia in pediatric patients: the UK experience

P O'Hare; Giovanna Lucchini; Michelle Cummins; Paul Veys; Michael Potter; Sarah Lawson; Ajay Vora; Rob Wynn; A Peniket; Keiren Kirkland; Rachel M. Pearce; J Perry; Persis Amrolia

We report outcomes for 44 children who underwent stem cell transplantation (SCT) for refractory AML in the UK between 2000 and 2012. Median age at SCT was 11.5 years. Twenty-three patients had primary refractory and 21 relapsed refractory AML. Refractory disease was confirmed by cytogenetics/molecular genetics in 24 cases. Median follow-up of the whole cohort is 6.8 years (2.1–14.9 years). Thirty patients (68%) achieved a CR following SCT. Transplant-related mortality at 1 year was 18%. Acute GVHD incidence was 52% (grade ⩾III 19%), chronic 7%. Relapse was the major cause of treatment failure and occurred in 32% of patients at a median of 61 days post SCT. Five-year overall survival and leukemia-free survival (LFS) were 43% (95% CI 31–61%). All patients with favorable cytogenetics (n=6) are alive in CR. Outcomes in patients with primary refractory disease were equivalent to those with relapsed refractory AML. Blast percentage ⩽30% in the BM pre-SCT, myeloablative conditioning and acute GVHD proved to be favorable prognostic features. We could stratify patients according to age ⩾10 years and >30% blasts in BM pre-SCT. Patients with none/one of these risk factors were highly salvageable (5 years LFS 53%) whereas those with both factors had a very poor prognosis (5 years LFS 10%). This may facilitate decision making on whether it is appropriate to consider transplant in such patients.

Collaboration


Dive into the Giovanna Lucchini's collaboration.

Top Co-Authors

Avatar

Paul Veys

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Persis Amrolia

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Kanchan Rao

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Robert Chiesa

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Peter Bader

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Austen Worth

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Juliana Silva

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Stuart Adams

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

Andrea Jarisch

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Jan Soerensen

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge