33 results on '"Simona Bernardi"'
Search Results
2. A Systematic Review of the Literature and Perspectives on the Role of Biomarkers in the Management of Malnutrition After Allogeneic Hematopoietic Stem Cell Transplantation
- Author
-
Marco Andreoli, Enrico Morello, Milena Giovanna Guarinoni, Michele Malagola, Alessandro Turra, Domenico Russo, Francesco Arena, Simona Bernardi, and Nicola Polverelli
- Subjects
lcsh:Immunologic diseases. Allergy ,medicine.medical_treatment ,Immunology ,Graft vs Host Disease ,chemical and pharmacologic phenomena ,Hematopoietic stem cell transplantation ,Bioinformatics ,Immune Dysfunction ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,immune system diseases ,graft versus host disease ,Humans ,Transplantation, Homologous ,Immunology and Allergy ,Medicine ,malnutrition (MeSH) ,business.industry ,Malnutrition ,insulin growth factor-1 (IGF-1) ,Hematopoietic Stem Cell Transplantation ,medicine.disease ,Graft-versus-host disease ,surgical procedures, operative ,030220 oncology & carcinogenesis ,citrulline ,Biomarker (medicine) ,biomarker ,Systematic Review ,business ,Gastrointestinal function ,lcsh:RC581-607 ,Biomarkers ,030215 immunology ,Systematic search - Abstract
Malnutrition is a common problem after allogeneic hematopoietic stem cell transplantation (allo-HSCT) and could impair immune function. Immune dysfunction after allo-HSCT may be linked with infections, GVHD, and relapse and negatively affect the outcome. Aim of this review was to identify malnutrition biomarkers, potentially useful for immune-system monitoring, in the setting of allo-HSCT. After a systematic search, no satisfying biomarker was found, except for citrulline. Citrulline could be useful in monitoring gastrointestinal function after allo-HSCT and its role in the complex relationship with immune-system function ought to be better explored. A multi-omics approach, including biomarkers and PRO (patient reported outcomes) is, in our opinion, the optimal way to study the relationship between malnutrition and transplant outcomes.
- Published
- 2021
- Full Text
- View/download PDF
3. Bioengineered scaffolding for mandibular reconstruction: a preclinical, xenograft animal study
- Author
-
Simona Bernardi, Marco Ferrari, R. Gilbert, P. Nicolai, J. Irish, Elisa Borsani, Kamol Dey, Luciana Sartore, Sowmya Viswanathan, D. Eu, D. Mattavelli, Smitha Mathews, T. Gualtieri, J. Townson, Federica Re, H. Chan, S. Taboni, Domenico Russo, and A. Sahovaler
- Subjects
Orthodontics ,Cancer Research ,Transplantation ,Scaffold ,business.industry ,Immunology ,Cell Biology ,Oncology ,Immunology and Allergy ,Medicine ,Animal study ,Mandibular reconstruction ,business ,Genetics (clinical) - Published
- 2021
4. Case Report: Late Onset of Myelodysplastic Syndrome From Donor Progenitor Cells After Allogeneic Stem Cell Transplantation. Which Lessons Can We Draw From the Reported Case?
- Author
-
Lisa Gandolfi, Tatiana Zollner, Mirko Farina, Alessandro Turra, Enrico Morello, Michele Malagola, Federica Cattina, Benedetta Rambaldi, Simona Bernardi, Doriana Gramegna, Domenico Russo, Nicola Polverelli, and Camilla Zanaglio
- Subjects
0301 basic medicine ,Cancer Research ,Monosomy ,medicine.medical_treatment ,Case Report ,immunosurveillance ,leukemogenesis ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,stem cells ,hemic and lymphatic diseases ,medicine ,Genetic predisposition ,transplant ,Progenitor cell ,immunosuppression ,business.industry ,Myelodysplastic syndromes ,Immunosuppression ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Transplantation ,Haematopoiesis ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Stem cell ,business ,donor cell myelodysplasia - Abstract
Background Myelodysplastic syndromes and acute leukemias after allogeneic stem cell transplantation (allo-SCT) are mainly caused by recurrence of the primitive leukemic clones. More rarely, they originate from donor hematopoietic stem cells, developing the so-called donor cell leukemia (DCL) or myelodysplastic syndromes (DC-MDSs). DCL and DC-MDS can be considered as an in vivo model of leukemogenesis, and even if the pathogenetic mechanisms remain speculative, a genetic predisposition of donor progenitor cells, an altered host microenvironment, and the impairment of immune surveillance are considered the main causes. Case presentation We report a case of DC-MDS diagnosed 5 years after an allo-SCT from a matched related donor (patient's sister) in a patient with Philadelphia chromosome-positive B-cell acute lymphoblastic leukemia (Ph+ B-ALL). The sex-mismatch allowed us to identify the donor cell origin. At the onset, the DC-MDS was characterized by chromosome seven monosomy and NRAS, RUNX1, and BCOR mutations. Because of a familiar history of colorectal neoplasia and the variant allele frequency (VAF) of NRAS mutation at the onset, this mutation was searched on germline DNA in both the donor and the recipient, but the result was negative. Moreover, after transplant (+4 months), the patient developed severe and long-lasting chronic graft-versus-host disease (cGVHD), requiring multiple lines of treatments. Because of the severe immunosuppression, recurrent infections occurred and, lately, the patient died due to septic shock. Conclusion This case report highlights the need, whenever possible, to evaluate the donor origin of the posttransplant myelodysplasia and acute leukemias. The potential key role of the impaired immune surveillance and of long-lasting immunosuppression appears to be emerging in the development of this case of DC-MDS. Finally, this case reminds the importance to investigate the familiar genetic predisposition in donors with a familiar history of neoplasia.
- Published
- 2020
- Full Text
- View/download PDF
5. Severe Acute Respiratory Syndrome Coronavirus-2 Pandemia: Facts and Perspectives in a Bone Marrow Transplant Unit
- Author
-
Federica Re, Tatiana Zollner, Lisa Gandolfi, Alessandro Turra, Domenico Russo, Camilla Zanaglio, Nicola Polverelli, Alessandro Isidori, Enrico Morello, Michele Malagola, and Simona Bernardi
- Subjects
Cancer Research ,2019-20 coronavirus outbreak ,haploidentical donor ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Bone marrow transplant unit ,allogeneic stem cell transplanation ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,donor search algorithm ,lcsh:RC254-282 ,Haploidentical Donor ,COVID 19 ,unrelated donor ,Oncology ,Unrelated Donor ,Immunology ,Medicine ,business - Published
- 2020
- Full Text
- View/download PDF
6. Successful hematopoietic stem cell transplantation for complete CTLA-4 haploinsufficiency due to a de novo monoallelic 2q33.2-2q33.3 deletion
- Author
-
Marcho Chiarini, Laura Palumbo, Luisa Gazzurelli, Nicola Polverelli, Daniele Moratto, Alessandro Turra, Alessio Benvenuto, Antonella Meini, Domenico Russo, Maria Federica Girelli, Simona Bernardi, Alessandro Plebani, Vassilios Lougaris, Enrico Morello, Michele Malagola, and Manuela Baronio
- Subjects
CTLA-4 ,HSCT ,medicine.medical_treatment ,Immunology ,Cancer research ,medicine ,Immunology and Allergy ,Hematopoietic stem cell transplantation ,Biology ,Haploinsufficiency - Published
- 2020
7. 3D-mapping of mesenchymal stem cells growth on bioengineered scaffolds for maxillofacial skeleton regeneration: a preclinical, in vitro study
- Author
-
Kamol Dey, T. Gualtieri, Elisa Borsani, D. Mattavelli, Luciana Sartore, Domenico Russo, Simona Bernardi, D. Eu, Smitha Mathews, Sowmya Viswanathan, J. Irish, Marco Ferrari, J. Townson, P. Nicolai, H. Chan, R. Gilbert, A. Sahovaler, Federica Re, and S. Taboni
- Subjects
Cancer Research ,Transplantation ,Regeneration (biology) ,Immunology ,Mesenchymal stem cell ,Cell Biology ,Biology ,Skeleton (computer programming) ,Cell biology ,3d mapping ,Oncology ,Immunology and Allergy ,In vitro study ,Genetics (clinical) - Published
- 2021
- Full Text
- View/download PDF
8. Comparative Somatic Mutational Profiling of CD34+ Hematopoietic Precursors (HSC) and Circulating Endothelial Cells (CEC) in Patients with Primary Myelofibrosis (PMF)
- Author
-
Mirko Farina, Ross L. Levine, Camilla Zanaglio, Andrew Dunbar, Nicola Polverelli, Michele Malagola, Giuseppe Rossi, Domenico Russo, Mariella D'Adda, Federica Cattina, Tatiana Zollner, Simona Bernardi, and Federica Re
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,education ,Immunology ,CD34 ,Cell Biology ,Hematology ,Gene mutation ,medicine.disease ,Biochemistry ,Haematopoiesis ,Immunophenotyping ,Specimen collection ,Internal medicine ,medicine ,In patient ,Sample collection ,business ,Myelofibrosis - Abstract
INTRODUCTION Endothelial cells (EC) play an important role in thrombosis and are increased in frequency in Myeloproliferative Neoplasms (MPN). Notably, the JAK2V617F mutation has been identified in micro-laser-dissected EC in peripheral vasculature (Sozer, 2009) and is present in a subset of endothelial progenitor cells (Teofili, 2011). Primary myelofibrosis (PMF) is also characterized by increased bone marrow vascularity. These data suggest that PMF clones could derive from a common precursor shared between CD34-hematopoietic stem cells (HSC) and EC. We explored this hypothesis with a prospective study aimed at comparing the mutational profiles of paired Circulating Endothelial Cells (CEC) and HSC in patients (pts) with PMF. METHODS 14 PMF pts not previously treated with JAK2 inhibitors, along with one healthy control, were enrolled in the MyCEC0617 study beginning in 2018. The study was approved by the local Ethical Committee and all pts gave written informed consent. HSC were selected using CD34+ immunomagnetic bead-column separation. CEC were detected using the CellSearch system, which uses immunomagnetic selection incorporating ferrofluid nanoparticles and fluorophore-labelled antibodies. The markers CD146, CD105, CD45, and DAPI were used to isolate CEC. We identified cells as CEC when we observed characteristic morphological features and a surface immunophenotype of CD146+, CD105 +, DAPI+ and CD45-. Putative CEC were then sorted using the DEPArray system, which uses a combination of di-electrophoresis technology and high-quality image-based cell selection to manipulate individual cells. Sequencing data was then assessed with the MiSeq Illumina NGS platform using a 54-gene custom panel focused on genes mutated in PMF. The cutoffs to confirm the presence of the mutations were identification of mutant alleles in 30 and 50 reads both in forward and reverse, for HSC and CEC, respectively. RESULTS The characteristics of pts are reported in Figure 1. Median age of pts was 69.5 ys (35-85) and male/female ratio was 9:6. Median time from diagnosis to sample collection was 4 (1-211) months. 2/14 pts had had a previous thrombosis, 78% had splenomegaly, and 29% presented with constitutional symptoms. Considering the molecular profile, 9 pts were JAK2V617F mutated, 3 were CALR mutated, and 1 was MPLW515L positive. 2 pts were triple-negative. Most of the pts (7) presented with an Intermediate-1 DIPSS score, while 5 were intermediate-2, and 2 were high risk DIPSS. CEC were collected for 12 of 15 subjects (including the normal control). No significative differences were found between pts from whom we isolated CEC and those we did not successfully recover CEC. A median of 26 (1-122) CEC in 4 ml of peripheral blood were recovered. Notably, no mutations were found in the CEC or HSC from healthy control whereas molecular alterations were discovered both on CEC and HSC in 11 pts (Fig. 1). The previously-identified MPN driver mutation was identified in MF HSC (except for one JAK2-mutated pt and for all CALR-mutated pts, consistent with the limitations of NGS in detecting these lesions). Interestingly, PMF pts demonstrated both shared and different mutations when comparing mutational profiles of HSC and CEC. In particular, 8 of 11 pts shared at least one mutation between EC and HSC. 2 pts harbored the same driver mutation (JAK2V617F), together with ABL1, IDH1, TET2, and ASXL1, respectively. The most frequently mutated genes shared between both CEC and HSC were JAK2, ASXL1, TET2, NOTCH1, and SRSF2. All of these mutations were observed as shared clonal events in at least 2 cases. We also identified individual paired HSC/CEC with shared mutations in TP53, KIT, KMT2A, IDH1, WT1 and ABL1. One pt shared 4 mutations between HSC and CEC, while 4 and 2 pts shared 1 and 2 mutations, respectively. CONCLUSION HSC and EC in PMF have in common one or more gene mutations implicated in the pathogenesis of PMF. For the first time, we show that mutations other than JAK2 can be identified in EC, and in most instances (70% of pts), these mutations are shared between CEC and HSC. Moreover, we present an analysis of mature cells of endothelial lineage including to the CEC. These preliminary findings using primary pts samples support the notion that PMF-initiating cells may be derived from a common HSC/EC precursor. Further data are needed to validate these findings and provide a rationale for additional studies exploring the antecedent cell of origin in MPN. Disclosures D'Adda: Pfizer: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Incyte: Membership on an entity's Board of Directors or advisory committees. Rossi:Celgene: Membership on an entity's Board of Directors or advisory committees; Amgen: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Jazz: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria; Mundipharma: Honoraria; BMS: Honoraria; Sandoz: Honoraria; Daiichi-Sankyo: Consultancy; Roche: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Levine:Qiagen: Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy; Loxo: Membership on an entity's Board of Directors or advisory committees; Roche: Consultancy, Research Funding; Prelude Therapeutics: Research Funding; Isoplexis: Membership on an entity's Board of Directors or advisory committees; C4 Therapeutics: Membership on an entity's Board of Directors or advisory committees; Imago Biosciences: Membership on an entity's Board of Directors or advisory committees; Gilead: Consultancy; Lilly: Honoraria; Amgen: Honoraria; Celgene: Consultancy, Research Funding.
- Published
- 2019
9. Multidimensional Geriatric Assessment for Elderly Patients (≥60 years) Submitted for Allogeneic Stem Cell Transplantation. a French (Paris) - Italian (Brescia) 10-Years Experience on 228 Patients
- Author
-
Federica Re, Enrico Morello, Michele Malagola, Giorgia Battipaglia, Simona Bernardi, Nicola Polverelli, Alessandro Turra, Domenico Russo, Paolo Tura, Gandolfi Lisa, Tatiana Zollner, Camilla Zanaglio, and Mohamad Mohty
- Subjects
medicine.medical_specialty ,Univariate analysis ,business.industry ,Incidence (epidemiology) ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Comorbidity ,Transplantation ,Log-rank test ,Internal medicine ,Cohort ,Population study ,Medicine ,business ,Cause of death - Abstract
Introduction. A multidimensional geriatric assessment has been shown to predict therapy-related toxicities in elderly hematology patients. No data are yet available on the use of combined functional and comorbidity scores in the allogeneic stem cell transplant (alloSCT) setting. The Lymphoma Italian Foundation (FIL) score, a multidimensional approach, combines the geriatric comorbidity score using the cumulative illness rating scale-geriatric (CIRS-G) with the activities of daily living (ADL) score, instrumental activity of daily living (IADL) score, and older age. Aims. The aim of the present study was to evaluate the role of the multidimensional FIL score in predicting alloSCT outcome, in terms of transplant-related mortality (TRM), relapse incidence (RI) and overall survival (OS). Patients and methods. Overall, 228 patients older than 60 years were submitted for alloSCT in Italy and France, during the last 10 years (2009-2018). Patients' characteristics are detailed in Table 1. The median age of the study population was 64 years (range, 60-76), and 155 (68%) were male. Acute myeloid leukemia was the most common indication for transplant (53%) and complete remission status was recorded in 54% of cases at transplantation. A matched unrelated donor, sibling donor or alternative donor was utilized in 41%, 31%, and 28% of cases, respectively. Stem cell source was derived from peripheral blood in 82%, bone marrow in 8%, and cord blood in 10% of cases. Utilization of a myeloablative conditioning regimen was reserved for only 20% of patients. Median number of CD34+ x10^6/Kg infused cells was 5 (range, 0,01-12,9), and of CD3+ x10^7/Kg cells 18 (range, 0,2-47,8). The median follow-up period of the cohort was 36 months (range, 12-138). Overall survival was estimated by the Kaplan-Meier method from the date of transplant to the date of last follow-up or death; the log-rank test was used to detect differences between subgroups. The Fine and Gray competing risk regression model was used for calculation of TRM and RI. Univariate and multivariate analyses were carried out using the Cox proportional-hazards regression model. Results. A total of 121 (53%) patients died at last follow-up. The cause of death was related to TRM in 60 (49%) cases comprising infection (31), toxicity (13), acute GVHD (9), and chronic GVHD (7); disease relapse was the ultimate cause of death in 61 (51%) cases . The 2-year expected TRM, RI and OS were 25%, 36% and 49%, respectively. The FIL score was measured in 215 patients and classified patients as 'fit' in 125 (58%) casesa nd 'unfit/frail' in 90 (42%). According to the FIL score, fit patients were more frequently in complete remission (CR) at alloSCT compared with unfit/frail patients (78% vs 21%, p2 (34%). Interestingly, the HCI-CI (HR 1.06, 95%CI 0.96-1.16, p=0.27) failed to predict alloSCT outcome. In contrast , FIL score, predicted fit patients to have a better 2- and 5-year OS of 66% and 59% compared to 32% and 30% in unfit/frail patients (p 80 (0.53, 95%CI 0.36-0.78, p=0.0013) and FIL frail/unfit status (2.83, 95%CI 1.93-4.17, p80 (HR 0,56, 95%CI 0,37-0,85, p=0.0066) and FIL score (HR 2,20, 95%CI 1,27-3,78, p=0.0046) maintained significant associations with OS (Figure 2). Conclusions. In a fairly large cohort of elderly patients (≥60 years) undergoing alloSCT at two European transplant centers, a multidimensional geriatric approach appears to be more accurate compared to a sole comorbidity assessment, in predicting alloSCT outcome and identifying patients at high risk of TRM. This simple tool could easily be applied before transplant for better patient selection and tailoring of treatment. Disclosures Mohty: Jazz Pharmaceuticals: Honoraria, Research Funding.
- Published
- 2019
10. First Interim Report of the Italian Multicentric Phase-III Randomized Study to Optimize TKIs Multiple Approaches - (OPTkIMA) in Elderly Patients (older than 60 years) with Ph+ Chronic Myeloid Leukemia (CML) and MR3.0/ MR4.0 Stable Molecular Response
- Author
-
Simona Bernardi, Sabina Russo, Domenico Russo, Francesca Lunghi, Monia Lunghi, Ferrari Maria Luisa, Mariella D'Adda, Fabio Efficace, Giovanna Rege Cambrin, Antonella Russo Rossi, Lara Aprile, Emilio Usala, Michele Baccarani, Maria Rosaria Coppi, Serena Rupoli, Bruno Martino, Cristina Bucelli, Gianantonio Rosti, Valentina Giai, Rosaria Sancetta, Gianni Binotto, Lisa Gandolfi, Monica Crugnola, Michele Malagola, Alessandra Iurlo, Mario Tiribelli, Nicola Polverelli, Mirko Farina, Atelda Romano, Fausto Castagnetti, Massimiliano Bonifacio, Antonietta Falcone, Fabio Stagno, Camilla Zanaglio, Micaela Bergamaschi, and Elisabetta Abruzzese
- Subjects
Oncology ,medicine.medical_specialty ,Intention-to-treat analysis ,business.industry ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,law.invention ,Dasatinib ,Imatinib mesylate ,Randomized controlled trial ,Nilotinib ,law ,Internal medicine ,Molecular Response ,medicine ,business ,Adverse effect ,medicine.drug - Abstract
Therapy with Tyrosine Kinase Inhibitors (TKIs) changed the fate of Philadelphia-positive Chronic Myeloid Leukemia (CML). At present, the therapeutic strategy aims to improve the management of the disease and the quality of life of the patients. In July 2015, we started a prospective multicentric randomized trial with the aim to validate the policy of the intermittent de-escalation treatment and to explore the impact of this strategy on the Quality of Life. To this purpose, CML patients older than 60 years in stable (≥2 years) MR3.0 or MR4.0 molecular response were randomized to receive a FIXED intermittent TKIs regimen (one month ON and one month OFF), as previously published (Russo D, Blood 2013; Russo D, BCJ 2015), versus a PROGRESSIVE intermittent TKIs regimen (one month ON and one month OFF for the 1st year; one month ON and two months OFF for the 2nd year; one month ON and three months OFF from the 3rd year) (OPTkIMA study, ClinicalTrials.gov: NCT02326311). Molecular monitoring was performed according to the 2015 ELN guidelines, every 3 months by RT-PCR on peripheral blood (Baccarani M,Blood 2013). In case of MR3.0 (MMR) loss, checked in two monthly consecutive RT-PCR analysis, patients were planned to exit the study and to resume TKIs daily. This first interim report have been focused on the patients who, by intention to treat, have completed the first year of the study for an historical comparison with the previous INTERIM trial (Russo D, Blood 2013; Russo D, BCJ 2015). During the first year, both the patients randomized in the FIXED and in the PROGRESSIVE arms were given the TKIs treatment one month ON and one month OFF. Up to June 2018, 177 patients have been enrolled by 26 Italian Hematological Centers (first patient randomized in July 2015) and 121/177 patients (68%) completed the first year of OPTkIMA study. The median age was 71 years (range 60-89) and 64% of the patients were belonging to the Sokal intermediate/high risk goup. 96/121 (79%), 14/121 (12%) and 11/121 (9%) patients were receiving imatinib (IMA), nilotinib (NILO) and dasatinib (DAS), at the time of enrollment. Overall, 59/121 (49%) and 62/121 (51%) patients have been randomized in the FIXED and PROGRESSIVE arm, respectively. 41/62 patients (66%) randomly assigned to the PROGRESSIVE arm have entered the second year of therapy. 34/121 patients (28%) went out of the study during the first year. The reasons for protocol discontinuation were: informed consent withdrawn (2 cases), second cancer (4 cases), loss of MR3.0 (28 cases). (Table 1). Among the 28 patients who lost the MMR, 17 and 11 were in MR4.0 and MR3.0, respectively, when they were enrolled into the study. Thus the probability of loosing the MR3.0 while on OPTkIMA was 21,7% at one year (Figure 1). All the 28 patients resumed TKIs continuously and all obtained at least the MR3.0 response, within 6 months and are currently included in the study follow up. The intermittent treatment was well tolerated, with 4 serious adverse events (1 appendicitis, 1 atrial fibrillation, 1 cardiac failure, 1 hip fracture) and 3 adverse events (1 diarrohea, 1 pruritus and 1 fever), none of which have been considered treatement-related. None of the patients experienced the TKI withdrawn syndrome. According to this first interim report, we found that a policy of intermittent TKIs administration in elderly patients is safe and well tolerated. Analysis of patient-reported QoL outcomes is ongoing and will further add information on the overall treatment effectivness of the new PROGRESSIVE intermittent TKI administration. After the 1st year, 28/121 patients (23%) lost MR3.0 and all of them re-gained the major molecular response within 6 months from resumption of continuous treatment. The probability of MR3.0 loss while on OPTkIMA at 1 year was 21,7% and this is quite comparable with the 20% MR3.0 loss observed in the previous INTERIM trial (Russo D, Blood 2013; Russo D, BCJ 2015). Disclosures Efficace: Bristol Meyers Squibb: Consultancy; Seattle Genetics: Consultancy; Lundbeck: Research Funding; TEVA: Research Funding; AMGEN: Research Funding; Incyte: Consultancy; Amgen: Consultancy; TEVA: Consultancy; Orsenix: Consultancy. Abruzzese:Novartis: Consultancy; BMS: Consultancy; Pfizer: Consultancy; Ariad: Consultancy. Bonifacio:Incyte: Consultancy; Pfizer: Consultancy; Amgen: Consultancy; Novartis: Research Funding; Bristol Myers Squibb: Consultancy. Castagnetti:Incyte: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Bristol Meyers Squibb: Consultancy, Honoraria; Novartis: Consultancy, Honoraria. Giai:Novartis: Consultancy; Pfizer: Consultancy.
- Published
- 2018
11. Comparative study on ATG-thymoglobulin versus ATG-fresenius for the graft-versus-host disease (GVHD) prophylaxis in allogeneic stem cell transplantation from matched unrelated donor: a single-centre experience over the contemporary years
- Author
-
Camilla Zanaglio, Valeria Cancelli, Francesca Schieppati, Nicola Polverelli, Marco Chiarini, Alessandro Turra, Viviana Giustini, Enrico Morello, Michele Malagola, Simone Perucca, Domenico Russo, Federica Cattina, Benedetta Rambaldi, Alessandra Sottini, Cristina Skert, Alessandro Montanelli, Simona Bernardi, and Luisa Imberti
- Subjects
endocrine system ,Cancer Research ,Thymoglobulin ,business.industry ,Retrospective cohort study ,Hematology ,Matched Unrelated Donor ,medicine.disease ,Transplantation ,03 medical and health sciences ,surgical procedures, operative ,0302 clinical medicine ,Graft-versus-host disease ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Gvhd prophylaxis ,Stem cell ,business ,Survival rate ,030215 immunology - Abstract
Two different rabbit anti-thymocyte globulin (ATG) formulations, thymoglobulin-ATG (tATG) and fresenius-ATG (fATG) are usually employed as prophylaxis for graft-versus-host-disease (GVHD), one of t...
- Published
- 2018
12. Single Step Multiple Genotyping by MALDI-TOF Mass Spectrometry, for Evaluation of Minor Histocompatibility Antigens in Patients Submitted to Allogeneic Stem Cell Transplantation from HLA-Matched Related and Unrelated Donor
- Author
-
Bruno Martino, Vilma Mantovani, Giuseppe Console, Simona Bernardi, Giovanni Martinelli, Domenico Pastore, Eleonora Toffoletti, Federica Cattina, Michele Malagola, and Alessandra Santoro
- Subjects
0301 basic medicine ,Human leukocyte antigen ,Stem cell transplantation, HLA, minor Histocompatibility antigens ,Article ,03 medical and health sciences ,immune system diseases ,hemic and lymphatic diseases ,Minor histocompatibility antigen ,Medicine ,Genotyping ,HLA ,Stem cell transplantation ,minor Histocompatibility antigens ,stem cell transplantation ,minor histocompatibility antigens ,business.industry ,lcsh:RC633-647.5 ,Myeloid leukemia ,Hematology ,lcsh:Diseases of the blood and blood-forming organs ,medicine.disease ,Minor Histocompatibility antigens ,Transplantation ,Leukemia ,030104 developmental biology ,Graft-versus-host disease ,surgical procedures, operative ,Immunology ,Stem cell ,business - Abstract
The outcome of patients underwent to allogeneic stem cell transplantation (allo- SCT) is closely related to graft versus host disease (GvHD) and graft versus leukemia (GvL) effects which can be mediated by mHAgs. 23 mHAgs have been identified and reported to be differently correlated with GVHD or GVL and the aim of this work was develop a method to genotype the mHAgs described so far. For this study we used MALDI-TOF iPLEX Gold Mass Array technology. We tested 46 donor/recipient matched pairs that underwent allo-SCT because of Philadelphia positive (Ph+) chronic myeloid leukemia (n = 29) or Ph+ acute lymphoblastic leukemia (n =17). Our data show that sibling pairs had a lesser number of mHAgs mismatches compared to MUD pairs. Notably, donor/recipient genomic mismatch on DPH1 was correlated with an increased risk of acute GvHD and LB-ADIR-1R mismatch on graft versus host direction was correlated with a better RFS with no increase of GvHD risk. Our work provides a simple, accurate and highly automatable method for mHAgs genotyping and suggest the role of mHAgs in addressing the immune reaction between donor and host.
- Published
- 2017
- Full Text
- View/download PDF
13. Sequential monitoring of lymphocyte subsets and of T-and-B cell neogenesis indexes to identify time-varying immunologic profiles in relation to graft-versus-host disease and relapse after allogeneic stem cell transplantation
- Author
-
Viviana Giustini, Michele Malagola, Nicola Polverelli, Federica Cattina, Simona Bernardi, Simone Perucca, Stefano Martellos, Marco Chiarini, Luisa Imberti, Claudia Ghidini, Alessandra Sottini, Cristina Skert, Valeria Cancelli, Alessandro Turra, Domenico Russo, Benedetta Rambaldi, Skert, Cristina, Perucca, Simone, Chiarini, Marco, Giustini, Viviana, Sottini, Alessandra, Ghidini, Claudia, Martellos, Stefano, Cattina, Federica, Rambaldi, Benedetta, Cancelli, Valeria, Malagola, Michele, Turra, Alessandro, Polverelli, Nicola, Bernardi, Simona, Imberti, Luisa, and Russo, Domenico
- Subjects
Genetics and Molecular Biology (all) ,Male ,B Cells ,Cell Transplantation ,T-Lymphocytes ,lcsh:Medicine ,Graft vs Host Disease ,Medicine (all) ,Biochemistry, Genetics and Molecular Biology (all) ,Agricultural and Biological Sciences (all) ,Signal transduction ,Biochemistry ,Neogenesis ,Memory T cells ,Steroid Therapy ,White Blood Cells ,0302 clinical medicine ,Mathematical and Statistical Techniques ,Animal Cells ,Recurrence ,Medicine and Health Sciences ,Blood and Lymphatic System Procedures ,Cytotoxic T cell ,Lymphocytes ,lcsh:Science ,B-Lymphocytes ,Multidisciplinary ,biology ,Pharmaceutics ,Hematopoietic Stem Cell Transplantation ,Middle Aged ,Flow Cytometry ,medicine.anatomical_structure ,surgical procedures, operative ,030220 oncology & carcinogenesis ,Physical Sciences ,Female ,Cellular Types ,Coreceptors ,Statistics (Mathematics) ,Research Article ,Adult ,Adolescent ,T cell ,Immune Cells ,Immunology ,T cells ,Surgical and Invasive Medical Procedures ,Cytotoxic T cells ,Research and Analysis Methods ,CD19 ,03 medical and health sciences ,Young Adult ,Drug Therapy ,medicine ,Humans ,Transplantation, Homologous ,Statistical Methods ,Antibody-Producing Cells ,B cell ,Aged ,Transplantation ,Blood Cells ,business.industry ,lcsh:R ,Biology and Life Sciences ,CD coreceptors ,Cell Biology ,biology.organism_classification ,medicine.disease ,Lymphocyte Subsets ,Graft-versus-host disease ,Multivariate Analysis ,biology.protein ,lcsh:Q ,business ,CD8 ,Mathematics ,030215 immunology ,Stem Cell Transplantation - Abstract
T and B lymphocyte subsets have been not univocally associated to Graft-versus-host disease (GVHD) and relapse of hematological malignancies after stem cell transplantation (SCT). Their sequential assessment together with B and T cell neogenesis indexes has been not thoroughly analysed in relation to these changing and interrelated immunologic/clinic events yet. Lymphocyte subsets in peripheral blood (PB) and B and T cell neogenesis indexes were analysed together at different time points in a prospective study of 50 patients. Principal component analysis (PCA) was used as first step of multivariate analysis to address issues related to a high number of variables versus a relatively low number of patients. Multivariate analysis was completed by Fine-Gray proportional hazard regression model. PCA identified 3 clusters of variables (PC1-3), which correlated with acute GVHD: PC1 (pre-SCT: KRECs≥6608/ml, unswitched memory B 44%, CD8+TCM cells>4%; HR 1.9, p = 0.01), and PC3 (at aGVHD onset: CD4+TEMRA69%, switched memory CD19+ = 0 cells and KRECs
- Published
- 2017
14. Detection of newly produced T and B lymphocytes by digital PCR in blood stored dry on nylon flocked swabs
- Author
-
Alessandra Sottini, Claudia Ghidini, Giovanni Martellosio, Federico Serana, Luisa Imberti, Marion Vaglio Tessitore, Aldo M. Roccaro, and Simona Bernardi
- Subjects
0301 basic medicine ,T-Lymphocytes ,lcsh:Medicine ,Digital PCR ,Flocked swabs ,KRECs ,TRECs ,Adolescent ,Adult ,Aged ,Aged, 80 and over ,B-Lymphocytes ,Blood Specimen Collection ,DNA ,Electrophoresis, Agar Gel ,HeLa Cells ,Humans ,Middle Aged ,Nylons ,Real-Time Polymerase Chain Reaction ,Recombination, Genetic ,Reproducibility of Results ,Young Adult ,0302 clinical medicine ,Agar Gel ,80 and over ,Digital polymerase chain reaction ,Statistical analysis ,Dried blood ,Immunodeficiency ,Medicine(all) ,General Medicine ,Real-time polymerase chain reaction ,030220 oncology & carcinogenesis ,Electrophoresis ,Peripheral blood mononuclear cell ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Genetic ,medicine ,In patient ,Biochemistry, Genetics and Molecular Biology(all) ,business.industry ,Research ,lcsh:R ,medicine.disease ,Virology ,DNA extraction ,Recombination ,030104 developmental biology ,Immunology ,business - Abstract
Background A normal number of T-cell receptor excision circles (TRECs) and K-deleting recombination excision circles (KRECs) is considered a biomarker for adequate new T- and B-cell production. In newborns, detection of TRECs and KRECs by real time PCR from dried blood spotted on filter paper is used for the screening of severe immunodeficiency. In adults, elderly and during diseases, where the number of TRECs is lower than in newborns and children, a large amount of DNA and a sensitive method of amplification are necessary to identify newly produced lymphocytes. Methods DNA was prepared from blood of 203 healthy adults (range: 18–91 years old) absorbed for 10 s on flocked swabs and let to dry, or from peripheral blood mononuclear cells. DNA was subjected to digital PCR and to well established conventional real time PCR-based method using TREC- and KREC-specific primers and probes. The number of TRECs and KRECs was expressed per mL of blood. Statistical analysis was performed by nested ANOVA, Pearson coefficient of determination, and by linear regression tests. Results The novel method for the storage of dried blood on nylon flocked swabs and the use of digital PCR allow quantification of TRECs and KRECs with high degree of sensitivity, specificity, accuracy, and precision. TRECs and KRECs were amplified by digital PCR in all tested blood samples, including those obtained from elderly individuals (>70 years old) and that were negative by real time PCR. Furthermore, values of TRECs and KRECs obtained by digital PCR were in the range of those acquired by real time PCR. Conclusions Our findings demonstrate that DNA isolation from dried blood on flocked swabs followed by digital PCR-based analysis represents a useful tool for studying new lymphocyte production in adults and elderly individuals. This suggests the potential use of the methodology when monitoring of clinical variables is limited by the number of molecules that can be amplified and detected, such as in patients with immunodeficiency or under immunosuppressive therapies. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1169-9) contains supplementary material, which is available to authorized users.
- Published
- 2017
15. Favourable/Intermediate ELN-Risk Acute Myeloid Leukemia to Transplant or Not to Transplant First-Line?
- Author
-
Valeria Cancelli, Domenico Russo, Enrico Morello, Michele Malagola, Benedetta Rambaldi, Simona Bernardi, Alesssandro Turra, Nicola Polverelli, and Federica Cattina
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,First line ,Complete remission ,Myeloid leukemia ,Newly diagnosed ,Transplant-Related Mortality ,03 medical and health sciences ,030104 developmental biology ,hemic and lymphatic diseases ,Internal medicine ,Immunology ,Medicine ,business ,Acute myeloid leukemias - Abstract
Favourable/intermediate ELN-risk acute myeloid leukemias (AMLs) (e.g. those harboring t(8;21) or inv(16) or NPM1A mutations or CEBP-alpha bi-allelic mutations) account for 30% to 50% of all newly diagnosed AMLs [1,2]. In this setting, conventional induction treatments may induce complete remission (CR) in up to 70% to 80%, but relapses still occur in 40% to 50% of cases and, at the end, no more than 30% to 40% of patients can be cured. Therefore, the optimization of post- remission therapy represents the greatest challenge in the treatment of favourable/intermediate-I ELN-risk AML.
- Published
- 2017
- Full Text
- View/download PDF
16. Digital PCR (Dpcr) a Step Forward to Detection and Quantification of Minimal Residual Disease (MRD) in Ph+/BCR-ABL1 Chronic Myeloid Leukemia (CML)
- Author
-
Camilla Zanaglio, Federica Cattina, Simone Perucca, Nicola Polverelli, Federica Re, Domenico Russo, ro Montanelli, Giuseppina Ruggieri, Aless, Simona Bernardi, Michele Malagola, and Valeria Cancelli
- Subjects
05 social sciences ,Myeloid leukemia ,Chromosomal translocation ,Imatinib ,Biology ,Blastic Phase ,medicine.disease ,Minimal residual disease ,03 medical and health sciences ,Haematopoiesis ,Leukemia ,0302 clinical medicine ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,0502 economics and business ,Immunology ,Cancer research ,medicine ,050211 marketing ,Stem cell ,medicine.drug - Abstract
Philadelphia-positive (Ph+), BCR-ABL1, chronic myeloid leukemia (CML) is a model of leukemia driven by a single, specific, chromosome translocation, the t (9;22) (q22;q11). This translocation, leading to a new, hybrid, leukemia-specific gene (BCR-ABL1) encoding for a deregulated tyrosine-kinase protein (p210), drives the leukemic transformation of hematopoietic stem cells [1-6] and induces the progression of the disease from the early chronic phase (CP) to the late blastic phase BP) which close the natural history of the disease. In the 2000s, the introduction of Imatinib, the first tyrosinekinase inhibitor (TKI) able to target the protein p210, significantly changed the fate of CML to fatal disease in real chronic disease.
- Published
- 2017
17. A specific Toll-like receptor profile on T lymphocytes and values of monocytes correlate with bacterial, fungal, and cytomegalovirus infections in the early period of allogeneic stem cell transplantation
- Author
-
E. Garrafa, A Di Palma, Rossella Ribolla, Cesare Bergonzi, Simone Perucca, Valeria Cancelli, Domenico Russo, Simona Bernardi, Arnaldo Caruso, Michele Malagola, Manuela Fogli, Cristina Skert, Alessandro Turra, Carla Filì, Federica Cattina, and Simona Fiorentini
- Subjects
Male ,Time Factors ,T-Lymphocytes ,Monocytes ,Killer Cells ,Interferon gamma ,Lymphocytes ,Prospective Studies ,Receptor ,Cells, Cultured ,Chemokine CCL2 ,Toll-like receptor ,Cultured ,Medicine (all) ,Toll-Like Receptors ,Stem cell transplantation ,Age Factors ,Bacterial Infections ,Middle Aged ,Killer Cells, Natural ,Survival Rate ,Infectious Diseases ,medicine.anatomical_structure ,Cytomegalovirus Infections ,Natural ,Female ,Tumor necrosis factor alpha ,medicine.drug ,Adult ,Homologous ,Adolescent ,Cells ,Infections ,Interferon-gamma ,Young Adult ,Immune system ,medicine ,Transplantation, Homologous ,Humans ,Lymphocyte Count ,Interleukin 4 ,Transplantation ,Tumor Necrosis Factor-alpha ,business.industry ,Monocyte ,Toll-Like Receptor 5 ,Mycoses ,Toll-Like Receptor 7 ,Toll-Like Receptor 9 ,Immunology ,Interleukin-4 ,business ,Toll-like receptors ,Stem Cell Transplantation - Abstract
Background Bacterial, fungal, and viral infections often affect non-relapse mortality after allogeneic stem cell transplantation (alloSCT). Recovery from infections depends on a balanced integration between innate and adaptive immune responses. In this complex interplay, a key role is played by Toll-like receptors (TLRs), which are sensors of pathogen-associated molecular patterns. To our knowledge, no previous study deals with both expression and function of all human TLRs together, in relation to infections in the setting of alloSCT. Methods We prospectively evaluated 9 TLRs by flow cytometry on T lymphocytes and monocytes of 35 patients in relation to infectious events from day +30 to day +120. Tumor necrois factor-alpha, interleukin-4, interferon-gamma, and monocyte chemoattractant protein-1 induction upon TLR activation was assessed by enzyme-linked immunosorbent assay on cell supernatants. Results In multivariate Cox regression analysis, levels of TLR-9 expression on T lymphocytes (P = 0.01) and values of natural killer cells (P = 0.01) correlated negatively with bacterial infections, whereas cytomegalovirus (CMV) infection resulted as a positive predictor. We observed a trend for negative correlation between TLR-7 levels on T lymphocytes and fungal infections (P = 0.07). Values of monocytes were negatively associated with CMV infection (P = 0.03), whereas levels of TLR-5 on T lymphocytes were positive predictors (P = 0.01). Age (P = 0.03) and bacterial infections (P = 0.006) negatively influenced overall survival. Monocyte values were positive predictors of survival (P = 0.003). Conclusions Bacterial, fungal, and CMV infections were associated with a different expression of some TLRs on T lymphocytes. The protective role of TLR-7 and TLR-9 seemed dominant over other TLRs involved in recognizing fungi and bacteria. We also observed an atypical involvement of TLR-5 in CMV infection. The dominant and atypical role of some TLRs could depend on their pleiotropic functions and the changing inflammatory environment of transplanted patients. A specific TLR profile and an adequate count of monocytes could improve survival, promoting an effective control of infections, and balanced immune responses. If our findings will be confirmed by further studies, these immunological variables could be useful as parameters to predict susceptibility to infections.
- Published
- 2014
- Full Text
- View/download PDF
18. Digital PCR (dPCR) Overcomes the Limitations in Detection and in Quantification of Quantitative PCR (qPCR) and Reveals Different Levels of BCR-ABL1 Copies/µl Among the Chronic Myeloid Leukemia (CML) Patients Achieving Major (MR3.0) or DEEP (MR4.0, MR4.5 and MR5.0) Molecular Response with Tyrosin Kynase Inhibitors (TKIs)
- Author
-
Simona Bernardi, Fausto Castagnetti, Maria Teresa Bochicchio, Valeria Cancelli, Andrea Di Palma, Simone Perucca, Crisitina Skert, Domenico Russo, Erika Codarin, Mario Tiribelli, Federica Cattina, Gianantonio Rosti, Michele Malagola, Luigi Caimi, Giuseppe Rossi, Giuseppina Ruggeri, Giovanni Martinelli, and Simona Bernardi, Andrea Di Palma, Mario Tiribelli, Erika Codarin, Giuseppina Ruggeri, Maria Teresa Bochicchio, Michele Malagola, Federica Cattina, Simone Perucca, Valeria Cancelli, Crisitina Skert, Gianantonio Rosti, Fausto Castagnetti, Luigi Caimi, Giuseppe Rossi, Giovanni Martinelli, Domenico Russo
- Subjects
Oncology ,Pathology ,medicine.medical_specialty ,Serial dilution ,business.industry ,Immunology ,Myeloid leukemia ,Imatinib ,Cell Biology ,Hematology ,Biochemistry ,law.invention ,Dasatinib ,DIGITAL PCR, CML, DEEP MOLECULAR RESPONSE ,Real-time polymerase chain reaction ,Nilotinib ,law ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Digital polymerase chain reaction ,business ,Polymerase chain reaction ,medicine.drug - Abstract
Monitoring BCR-ABL1 transcript by quantitative PCR (qPCR) is essential for the management of CML patients treated with TKIs. Currently, up to 40-50% of CML patients treated with TKIs can achieve a deep molecular response (DMR = BCR-ABL1 minor or equal to0,01% IS), but only 50% of them are reported to maintain a stable Treatment Free Remission (TFR). Since qPCR has some intrinsic limitations in detection and quantification of BCR-ABL1 transcript, this method does not appear optimal in the selection of patients eligible for TKIs cessation. A precise monitoring of BCR-ABL1 transcript levels can help even better the clinicians in managing CML patients treated with TKIs and in selecting the best candidates for discontinuation of TKIs without relapse. Digital PCR (dPCR) can be more advantageous than qPCR. It gives the absolute quantification of target nucleic acidsby partitioning the PCR reaction mix over a large number of wells, each containing a single copy or no copies of the target region. Based on the assumption of Poisson's distribution, the number of template copies originally presenting in the sample can be calculated from the number of partitions in which amplification has successfully occurred. In that way, standard curves cannot be necessary and data are more accurate. In this study we set a dPCR assay to quantify the BCR-ABL1 transcript in a preliminary cohort of CML patients with major (MMR or MR3.0) or deep molecular response (MR4.0, MR4.5 and MR5.0). The analysis by dPCR were based on a TaqMan-MGB probes targeting the BCR-ABL1 transcript. Custom assay was designed and produced with a FAM-label, basing on the sequence of routinely-used probes. The experiments were performed on a QuantStudio 3D Digital PCR System (Life Technologies). A commercial BCR-ABL1 Mbcr standard dilutions (Qiagen) and 10 replicates of blank samples (DNA-free, RNA-free water) were used to set dPCR assay and to determine the Limit of Detection and the Limit of Quantification of our method. The values of absolute quantities of BCR-ABL1 transcript assessed by dPCR were expressed as number of copies/ul. Samples for dPCR testing were obtained from peripheral blood (10 ml in EDTA tubes) of CML patients treated with TKIs, namely imatinib, nilotinib, or dasatinib, on time-checks planned for monitoring MMR or DMR through the conventional qPCR. To the purpose of the study, we evaluated 10 cases with stable MR3.0, 7 cases with stable MR4.0, 6 cases with stable MR4.5, 7 cases with stable MR5.0. It has been considered as stable any MR3.0, MR4.0, MR4.5, or MR5.0 molecular response measured by conventional qPCR and detected in the last three consecutive checks performed during the last 12 months. Blank samples served as negative controls, while 10 peripheral blood samples of healthy donors served as normal controls. Digital PCR (dPCR) revealed different levels of BCR-ABL1 copies/µl among the CML patients achieving the major (MR3.0) or deep (MR4.0, MR4.5 AND MR5.0) molecular response with TKIs. Moving from MR3.0 to MR5.0 molecular response the median of BCR-ABL1 copies/µl assessed by dPCR were progressively decreasing (Figure 1), with blanks and healthy controls approximately to zero. Medians with ranges were 0.957 (0.472-1.692) for MR3.0; 0.319 (0.072-0.906) for MR4.0; 0.231 (0.063-0.651) for MR4.5; 0.219 (0.074-0.399) for MR5.0; 0.076 (0.000-0.118) for healthy controls, and 0.000 (0.000-0.129) for blanks. Moreover, dPCR revealed different BCR-ABL1 copies/µl among the patients of each class of molecular response. Importantly, in patients with MR4.5, MR5.0 and with undetectable levels of BCR-ABL1 % IS as measured with qPCR, discrete variable levels of BCR-ABL1 copies/µl have been detected by dPCR. These data, revealing different levels of BCR-ABL1 copies/µl beyond the limit of detection and quantification of conventional qPCR, may explain why no correlation was observed between BCR-ABL1 % IS levels measured by qPCR and numbers of BCR-ABL1 copies/µl measured by dPCR. We are screening CML patients with MMR and DMR, but these preliminary results show that dPCR appears to be more accurate than qPCR for detection and quantification of BCR-ABL1 transcript and it should be seen as a useful step forward in order to better manage the TKI therapy and to better select the candidates for TFR. Acknowledgments: Department of Clinical and Experimental Sciences, University of Brescia; PRIN2009; European Leukaemia Net; BCC "Pompiano e Franciacorta". Figure 1. Figure 1. Disclosures Tiribelli: Bristol Myers Squibb: Consultancy, Speakers Bureau; Ariad Pharmaceuticals: Consultancy, Speakers Bureau; Novartis Farma: Consultancy, Speakers Bureau. Rosti:Bristol Myers Squibb: Honoraria, Research Funding, Speakers Bureau; Novartis: Honoraria, Research Funding, Speakers Bureau. Martinelli:Roche: Consultancy; BMS: Speakers Bureau; MSD: Consultancy; ARIAD: Consultancy; Novartis: Speakers Bureau; Pfizer: Consultancy.
- Published
- 2015
19. Aggressive Aneuploid Acute Myeloid Leukemia Is Dependent on Alterations of P53, Gain of APC and PLK1 and Loss of RAD50
- Author
-
Marianna Garonzi, Maria Chiara Fontana, Viviana Guadagnuolo, Antonella Padella, Carmen Baldazzi, Maddalena Raffini, Sara Bacilieri, Cristina Papayannidis, Nicoletta Testoni, Anna Maria Ferrari, Elisa Zuffa, Antonella Laginestra, Torsten Haferlach, Massimo Delledonne, Eugenia Franchini, Giovanni Martinelli, Daniel Remondini, Simona Bernardi, Annalisa Astolfi, Italo Faria do Valle, Samantha Bruno, Giovanni Marconi, Giorgia Simonetti, Alberto Ferrarini, Marco Manfrini, Emanuela Ottaviani, Simonetti, Giorgia, Padella, Antonella, Manfrini, Marco, FARIA DO VALLE, Italo, Papayannidis, Cristina, Fontana, MARIA CHIARA, Guadagnuolo, Viviana, Baldazzi, Carmen, Ferrari, Anna, Bruno, Samantha, Ferrarini, Alberto, Bernardi, Simona, Garonzi, Marianna, Astolfi, Annalisa, Marconi, Giovanni, Zuffa, Elisa, Franchini, Eugenia, Ottaviani, Emanuela, Laginestra, MARIA ANTONELLA, Raffini, Maddalena, Bacilieri, Sara, Testoni, Nicoletta, Delledonne, Massimo, Haferlach, Torsten, Remondini, Daniel, and Martinelli, Giovanni
- Subjects
0301 basic medicine ,Genetics ,Genome instability ,Monosomy ,Protein catabolic process ,Immunology ,Myeloid leukemia ,Aneuploidy ,Cell Biology ,Hematology ,Biology ,Cell cycle ,medicine.disease ,Biochemistry ,Protein ubiquitination ,Aneuploidy, acute myeloid leukemia ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,medicine ,Cancer research - Abstract
Chromosome number alterations, aneuploidy, is a hallmark of cancer. It occurs in about 15% of acute myeloid leukemia (AML) cases, is generally preserved throughout disease progression (Bochtler et al. Leukemia 2015) and correlates with adverse prognosis (Breems et al. JCO 2008, Papaemmanuil et al. NEJM 2016). This evidence highlights the need of understanding the molecular mechanisms that promote and sustain aneuploidy in AML, in order to define novel potential therapeutic targets. In the NGS-PTL project we profiled the genomic landscape of 536 hematological samples by whole exome sequencing (WES, Illumina). Among them, we analyzed 88 and 68 samples from aneuploid (A-) FLT3-wildtype AML (isolated trisomy and monosomy, complex and monosomal karyotype) and euploid (E-) AML (normal and complex karyotype, A-AML showed an increased genomic instability, as confirmed by a higher mutation load compared with E-AML (median number of variants: 22 (range: 2-95) and 11 (range: 3-45), respectively, pA substitutions, compared with the C>T transition-related signature, which is prevalent in AML. A-AML was associated with mutations and/or heterozygous deletion of TP53 (p We show here for the first time the molecular mechanisms promoting and maintaining aneuploidy in AML. Our results indicate that p53 deficiency, either caused by somatic mutations, copy number loss, impaired DNA damage response and enhanced PLK1 signaling synergize with APCgain, RAD50 structural or functional loss and forced progression through mitosis, to override cell cycle and mitotic checkpoints and allow the formation of daughter cells with an aberrant chromosome number. These mechanisms cooperate with recurrent mutations of genes involved in protein ubiquitination and proteasome-mediated protein catabolic process in A-AML, indicative of the attempt of aneuploid cells to override the proteotoxic stress due to the unbalanced protein load generated by the aneuploid condition. This evidence provides the rationale for exploiting proteasome inhibition (Velcade), p53 reactivation (MDM2/4 inhibitor) and targeting of the cell cycle (CHK1/2 inhibitor) downstream to p53 (WEE1 inhibitor) as strategies for novel combination therapies against aggressive aneuploid AML, which are under clinical investigation in our Institution and may serve as a model for aneuploid tumors. GS and AP: equal contribution. Supported by: FP7 NGS-PTL project, ELN, AIL, AIRC, PRIN, progetto Regione-Università 2010-12 (L. Bolondi). Disclosures Haferlach: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Martinelli:Ariad: Consultancy, Speakers Bureau; MSD: Consultancy; Celgene: Consultancy, Speakers Bureau; Roche: Consultancy, Speakers Bureau; Genentech: Consultancy; Novartis: Speakers Bureau; BMS: Speakers Bureau; Pfizer: Consultancy, Speakers Bureau; Amgen: Consultancy, Speakers Bureau.
- Published
- 2016
20. Comparative Study on Fresenius-ATG Versus Thymoglobuline-ATG for the Graft Versus Host Disease (GVHD) Prophylaxis in Allogeneic Stem Cell Transplantation from Matched Unrelated Donor: A Single Center Experience on 76 Patients
- Author
-
Alessandro Turra, Benedetta Rambaldi, Valeria Cancelli, Francesca Schieppati, Nicola Polverelli, Rossella Ribolla, Marta Bonetti, Domenico Russo, Cristina Skert, Simona Bernardi, Federica Cattina, Camilla Zanaglio, Michele Malagola, and Simone Perucca
- Subjects
medicine.medical_specialty ,Acute leukemia ,Hematology ,business.industry ,Incidence (epidemiology) ,Immunology ,Cell Biology ,Single Center ,medicine.disease ,Biochemistry ,Gastroenterology ,Transplantation ,Graft-versus-host disease ,Internal medicine ,Medicine ,Cumulative incidence ,Chills ,medicine.symptom ,business - Abstract
Introduction . Anti-thymocyte globulin (ATG), has been shown to significantly reduce the incidence and severity of acute and chronic graft versus host disease (GVHD) in pts submitted toallogeneic stem cell transplantation (allo-SCT). Two different ATG formulations, derived from rabbit immunization against T antigens,thymoglobuline-ATG (tATG) andfresenius-ATG (fATG) are usually employed. However, only few retrospective studies are available in order to compare the efficacy and safety of these two drugs. Moreover, the differential impact of the two formulations on immune recovery has been poorly studied. Methods. Clinical and laboratory data of 76 pts receivingfATG (30 mg/kg for 3 days beforeallo-SCT) ortATG (10 mg/Kg for 3 days beforeallo-SCT) as GVHD prophylaxis forallo-SCT from matched unrelated donor (MUD) for hematological malignancies at the Unit of Stem Cells Transplantation ofSpedaliCivili of Brescia between 2009 and 2014, were retrospectively collected. Basic phenotype of circulating lymphocytes was assessed by flow-cytometry at 3-months interval fromallo-SCT. The aims of the study were to compare the two ATG formulations in term of incidence of acute and chronic GVHD, infections, non-relapse mortality, relapse-free survival, overall survival and immune recovery. The study was approved by the local Ethic Committee. Results. Overall, 46 (60%) pts receivedfATG and 30 (40%)tATG as GVHD prophylaxis, in association to cyclosporine andmethotrexate. Baseline characteristics were (median): age, 46 years (range, 17-66); male, 68%; acute leukemia, 53%; complete hematological remission, 51%; HLA full-matched (8/8), 40%; source of stem cells, peripheral blood, 87%;myeloablative conditioning regimen, 49%; median follow-up, 22 months (range, 1-88). No significant differences were observed between the two populations according age, sex, hematological disease, disease status atallo-SCT, HLA match, stem cell source, amount of CD3+ and CD34+ cells infused and conditioning regimen. Overall, 26 (57%)fATGand 19 (61%)tATG-exposed pts developed an acute GVHD (p=0.8); among 18 (24%) pts who developedcGVHD, 2 out of 10 (20%)fATGand 6 out of 8 (75%)tATG-receiving pts evolved towards severecGVHD(p=0.05). In particular, the cumulative incidence of severecGVHD, was 5,2% versus 27,6% at 2 years (p=0.03) infATGandtATGgroup, respectively (Figure 1). Bacterial infections were encountered in 42 (91%) and 25 (81%) (p=0.19), fungal infections in 12 (26%) and 6 (19%) (p=0.58)fATGandtATG-treated pts. CMV reactivation was observed in 28 (61%) and 19 (73%) (p=0.43)fATGandtATG-exposed pts, respectively. However, a trend for a higher incidence of early-CMV reactivation was seen in thetATGgroup, with a cumulative incidence of CMV reactivation at 30 days of 30% versus 52% infATGandtATGcohort, respectively (p=0.09). No differences were observed betweenfATGandtATGpopulations in term of relapse-related mortality (24% versus 40%, p=0.25), non-relapse mortality (28% versus 20%, p=0.48) and overall survival (52% versus 60%, p=0.71). Immunologic reconstitution was evaluated in 48 pts (fATG, 64%). As compared totATG,fATGpts showed a significant lower amount of CD3-/CD16+ NK cells (median, 137/mclvs255/mcl, p=0.05) and a trend for lower CD3+CD4+CD25+CD127- T-regcells (5/mclvs11/mcl, p=0.07) and CD4/CD8 ratio (0.26vs0.57, p=0.08) at 3 months fromallo-SCT. At 6 months CD4/CD8 ratio was significantly lower infATGcompared totATGgroup (0.26vs0.42, p=0.01). At 9 months, lymphoid populations did not differ between the two cohorts. Overall, ATG administration was well-tolerated without grade III-IV adverse events, however an increase in infusion-related events (mainly, fever and chills) was recorded infATGgroup (67%vs32%, p=0.004). Conclusion. This retrospective study, reporting a quite large population ofallo-SCT pts from MUD homogeneously followed in a single Hematology Center, shows thatfATG-treated pts have a lower incidence of severecGVHD compared totATG. On the contrary a slight increase of early CMV reactivation has been observed intATG group. These differences do not result in a higher relapse-related or overall mortality. The distinct immune reconstitution as identified by flow-cytometricanalysis, could justify these observations, that need to be validated by prospective studies. Figure 1 Cumulative incidence of severecGVHD according to type of ATG employed Figure 1. Cumulative incidence of severecGVHD according to type of ATG employed Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
21. Comparative Monitoring of Minimal Residual Disease (MRD) By RT-Quantitative (RT-qPCR) and Digital PCR (dPCR) in Ph+ Chronic Myeloid Leukemia (CML) Patients Treated with TKIs for Recognition of Stable Deep Molecular Response (DMR) and Identification of Best Candidates to TKIs Treatment Discontinuation
- Author
-
Luca Franceschini, Massimiliano Bonifacio, Francesco Di Raimondo, Fabio Stagno, Simona Bernardi, Giuseppe Rossi, Camilla Zanaglio, Nicola Polverelli, Elisa Cerqui, Mirko Farina, Cristina Bucelli, Mario Tiribelli, Eleonora Toffoletti, Malgorzata Monika Trawinska, Marco Gobbi, Alessandra Iurlo, Michele Malagola, Gianluca Gaidano, Mariella D'Adda, Domenico Russo, Stefania Stella, Maria Teresa Voso, Micaela Bergamaschi, Clara Deambrogi, Elisabetta Abruzzese, Doriana Gramegna, Elif Dereli Eke, and Paolo Vigneri
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Immunology ,Disease progression ,Cell Biology ,Hematology ,Biochemistry ,Minimal residual disease ,Peripheral blood ,Prognostic score ,Discontinuation ,03 medical and health sciences ,Identification (information) ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,Digital polymerase chain reaction ,business ,Bristol-Myers - Abstract
Current therapy of CML with tyrosine kinase inhibitors (TKIs) aims to achieve a Major Molecular Response to avoid the disease progression to blastic phase and, possibly, a Deep Molecular Response (DMR), to gain the opportunity for TKIs discontinuation and Treatment Free Remission (TFR). However, TFR strategies are not yet optimized, as well as the criteria for patients (pts) selection to TKI discontinuation are not well defined. The patient's CML prognostic score, the duration of TKI treatment and "stable" DMR before discontinuation and the levels of DMR are still matters of debate. Due to its intrinsic limitations, RT-qPCR cannot be considered as an optimal tool for a precise detection of minimal BCR-ABL1 transcript levels and DMR monitoring. The digital PCR (dPCR) has emerged as a more sensitive and accurate method to detect and monitor the BCR-ABL1 minimal residual disease (MRD). This study focused on the BCR-ABL1 RT-q/dPCR comparative monitoring in CML pts treated with TKIs and with durable DMR (≥ 2 years), as conventionally assessed by RT-qPCR, before the enrollment. The aim was to evaluate the reliability and the efficiency of dPCR, as compared to RT-qPCR, for a better identification of "stable" DMR and for a better selection of the candidates for treatment discontinuation. 118 CML pts with durable DMR were enrolled and 493 peripheral blood samples were comparatively analyzed by both RT-qPCR, according to the last International Guidelines, and dPCR (QS3D Digital PCR System). RT-qPCR results were assessed as BCR-ABL1 %IS. dPCR results were assessed as number of BCR-ABL1 copies/□l of reaction and the molecular response was categorized as □ 0.468 or < 0.468 BCR-ABL1 copies/□l according to the previously reported cut off (Bernardi S. et al, J Mol Biom Diagn, 2017). Starting from the first RTq/dPCR comparative assessment, the 118 pts were monitored every 3/4 months for at least 3 time points. Grouping the 118 pts according to the RT-qPCR molecular results at the enrolment, 50 cases (42%) resulted in the MR4.0group and 68 (58%) in the MR 4.5-MR5.0 group. No significant differences were observed between these 2 groups in terms of clinical and hematological characteristics. At the same time, the 118 pts were divided in 2 groups based on the comparative dPCR molecular results, according to the above mentioned cut-off value: 29 (25%) and 89 (75%) cases had dPCR values □ and □0.468 BCR-ABL1 copies/□l, respectively. No significant clinical differences were observed between these 2 groups. Out of these 118 pts, 87 (74%) discontinued the TKI treatment and 24 (28%) of them lost the DMR after a median time of 3 months (range 1-30). Based on dPCR, 23/87 (26%) and 64/87 (74%) had dPCR □ 0.468 and < 0.468, respectively. Considering the pts who lost DMR, 12/23 (41%) and 12/64 (20%), had dPCR □ 0.468 and < 0.468, respectively (p=0.0021). Therefore, evaluating BCR-ABL1 transcript levels by dPCR before the discontinuation, we could observe a better TFR (positive predictive value of 80%) in those pts with dPCR < 0.468, with respect to the ones with □ 0.468. However, no significant difference was observed between the groups divided according to the evaluation of BCR-ABL1 transcript levels by RT-qPCR before the discontinuation in terms of TFR prediction. In order to reach a better recognition of stable DMR, which is strictly related with the probability to achieve and maintain TFR after TKIs discontinuation, we comparatively analyzed, by RTq/dPCR, the course of MRD during the follow up period. In the case of monitoring by RT-qPCR, a wide oscillation of the molecular responses was observed and they tended to be superimposable both in the group of pts with MR4.0 and in those with MR4.5 and 5.0. In the case of monitoring by dPCR, the values assessed at the enrolment and during the follow up revealed the capability of the dPCR to measure different and heterogeneous numbers of BCR-ABL1 copies in each pts. Moreover, the pts with a dPCR below the cut-off value of 0.468, at enrolment, were maintaining values (median 0,175; range 0-1,863), significantly lower (p This retrospective study show that the dPCR is more accurate and sensitive than conventional RT-qPCR for detecting and monitoring the BCR-ABL1 molecular levels and potentially able to improve the recognition of the stable DMR and selection of the candidates to TKI treatment discontinuation. Figure 1. Figure 1. Disclosures Bonifacio: Incyte: Consultancy; Pfizer: Consultancy; Amgen: Consultancy; Novartis: Research Funding; Bristol Myers Squibb: Consultancy. Gaidano:Morphosys: Honoraria; Amgen: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Gilead: Consultancy, Honoraria; Roche: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria. Rossi:Teva: Membership on an entity's Board of Directors or advisory committees; Mundipharma: Honoraria; Sanofi: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria; Gilead: Membership on an entity's Board of Directors or advisory committees; Sandoz: Honoraria; Jazz: Membership on an entity's Board of Directors or advisory committees; Roche: Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria; Amgen: Membership on an entity's Board of Directors or advisory committees. Gobbi:Novartis: Consultancy; Ariad: Membership on an entity's Board of Directors or advisory committees; Pfister: Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy; Janssen: Consultancy; Celgene: Membership on an entity's Board of Directors or advisory committees. Voso:Celgene: Research Funding, Speakers Bureau. Abruzzese:Ariad: Consultancy; BMS: Consultancy; Novartis: Consultancy; Pfizer: Consultancy.
- Published
- 2018
- Full Text
- View/download PDF
22. Minimal Residual Disease Detection at RNA and Leukemic Stem Cell (LSC) Level. Comparison of Qpcr, d-PCR and CD26 Stem Cell Measurements in Chronic Myeloid Leukemia (CML) Patients in Deep Molecular Response (DMR)
- Author
-
Camilla Zanaglio, Malgorzata Monika Trawinska, Simona Bernardi, Elisabetta Abruzzese, Francesco Bondanini, Michele Malagola, Paola Pacelli, Anna Sicuranza, Donatella Raspadori, Paolo de Fabritiis, and Monica Bocchia
- Subjects
education.field_of_study ,05 social sciences ,Immunology ,Population ,CD34 ,Cell Biology ,Hematology ,030204 cardiovascular system & hematology ,Biology ,Biochemistry ,Minimal residual disease ,Molecular biology ,Isotype ,Dasatinib ,03 medical and health sciences ,0302 clinical medicine ,Imatinib mesylate ,Nilotinib ,hemic and lymphatic diseases ,0502 economics and business ,medicine ,050211 marketing ,Digital polymerase chain reaction ,education ,medicine.drug - Abstract
Background. A Deep Molecular Response (DMR), defined as BCR-ABL1 p210 transcript at levels Aim. To compare and examine values of the three different methods of detecting minimal residual disease (MRD) in CML at RNA and LSC levels in patients in TFR or DMR. Patients and Methods. The twenty-seven patients in this study received treatment with either Imatinib (12), Dasatinib (6), Nilotinib (7), Bosutinib (1) or Interferon (1). Twelve patients were in TFR, while the rest were in DMR. TFR patients had stopped therapy for less than 1 year (3), 250,000 ABL copy numbers). D-PCR analysis used TaqMan-MGB probes targeting the BCR-ABL1 transcript. A custom assay was designed and produced with a FAM-label, basing on the sequence of routinely-used probes. BCR-ABL1 quantifications were performed analyzing 50ng of cDNA on a QuantStudio 3D Digital PCR System (ThermoFisher Scientific). BCR-ABL1 transcript values using dPCR were expressed as number of copies/ul. The secondary analysis were carried out by AnalysisSuite Cloud Software (ThermoFisher Scientific). To detect peripheral blood circulating CD34+/CD38-/CD26+ LSCs, cells were incubated with anti-CD45 (BD Biosciences), anti-CD34 (581), anti-CD38 (HIT2) and anti-CD26 (M-A261) (BD Pharmigen). Acquisition and analysis were performed by FACSCanto II flow cytometer (BD Biosciences, NR Nannini). CD45+ cells acquired for each sample ranged from 500,000 to 1,000,000. Isotype controls were included in each staining. Median absolute number of CD26+ cells/μL were calculated as follows: (# WBCs/μL) × (% CD34+/CD38−/CD26+ stained CD45+ cells). Results. TFR status, the type of TKI treatment, QPCR results, dPCR data and LSCs quantification are summarized in Table 1. Both d-PCR and LSCs showed higher sensitivity than QPCR, exhibiting positive results when transcript levels using QPCR were undetectable (16). None of the patients tested negative with d-PCR, however 14/16 were under the threshold of 0.468 copies/uL, corresponding to a stable DMR. LSC levels were negative in 5 patients, 3 of which also tested negative with QPCR. In all patients QPCR ranged from 0-0.0068, d-PCR from 0.073 to 0.943, and LSCs from 0 to 0.156. Results were divided in quartiles, depending on molecular response for QPCR, the 0.468 threshold for dPCR and the distribution for the LSCs. The 2 lowest quartiles defined the lowest detectable DMR. A strong correlation of these data in TFR patients was noted (10/12 concordant) while 8/15 DMR patients were discordant. Conclusions To our knowledge this is the first attempt to analyze and compare DMR in a CML population using standard (QPCR) and highly sensitive (dPCR and LSCs) methods. TFR patients, some lasting up to 17 years, were in the lowest detectable DMR categories. Very little is known about the biology of CML implications of circulating LSCs. Larger studies and dynamic scoring will help define their informative and predictive value. Disclosures Abruzzese: Pfizer: Consultancy; Ariad: Consultancy; Novartis: Research Funding; BMS: Consultancy.
- Published
- 2018
- Full Text
- View/download PDF
23. Identification of a Novel Mutation Predisposing to Familial AML and MDS Syndrome By a NGS Approach
- Author
-
Nicola Polverelli, Stefania Masneri, Mirko Farina, Federica Cattina, Domenico Russo, Simona Bernardi, Francesca Schieppati, Alessandro Turra, Benedetta Rambaldi, Camilla Zanaglio, Enrico Morello, Michele Malagola, and Elif Dereli Eke
- Subjects
0301 basic medicine ,Genetics ,Sanger sequencing ,Mutation ,Immunology ,MiRNA binding ,Cell Biology ,Hematology ,Biology ,medicine.disease_cause ,Biochemistry ,Genetic analysis ,Germline ,03 medical and health sciences ,symbols.namesake ,030104 developmental biology ,Germline mutation ,CEBPA ,symbols ,medicine ,Exome sequencing - Abstract
Introduction In AML and MDS cases, the genetic lesions inherited or acquired by the hematopoietic stem cells are considered as starting events. Familial AML and MDS, recently recognized in the revised WHO classification (2016) provide a useful model for investigation of predisposing genetic mutations. Genetic analysis of several pure familial leukemia pedigrees led to the discovery of well defined syndromes associated with inherited de novo mutations on germline DNA. Growing clinical awareness as well as a widespread use of NGS have led to an enlarged description of familial MDS/AML cases, and the number of mutations involved, suggesting they are more frequent than those previously recognized. Despite the recent discovery of well-established causative gene mutations (RUNX1, GATA2, ETV6, TERT, TERC, SRP72, ANKRD26, DDX41, CEBPA), many cases remain unexplained (about 80%), suggesting that other inherited mutations could predispose to MDS/AML. It is expected that new sequencing approaches will help to the identification of more cases, more genes as well as novel syndromes. In 2017, we started a multicentric prospective study (Clinical trial.gov NCT03058588) aiming to look for predisposing mutations in patients and relatives affected by Familial AML and MDS syndromes (FAMS) by NGS and to screen for old and new mutations potentially associated with the disease. Methods At present, 12 AML/MDS patients have been enrolled. Leukemic (bone marrow) and germline (buccal swab) DNA were analyzed by NGS gene panel approach based on a 28 genes associated to myeloid leukemogenesis, including the 9 above mentioned genes associated to FAMS. NGS libraries were performed by a Nimblegen (Roche) custom panel based on gene capture strategy and the sequencing was performed by MiSeq (Illumina). Results Ten patients did not reveal any germline mutations and the candidates are undergoing to whole exome sequencing. One presented a germline mutation on RUNX1, and the analysis of the affected relatives is on going. One revealed a new mutation. She was a 70 years old woman affected by RARS and her pedigree was characterized by 9 relatives affected by hematologic and solid neoplasia and trombocytopenia (fig 1). The NGS analysis revealed the mutation c.*514C>T in 3'UTR of ETV6 with VAF of 50% on tumor DNA. The variant has never been described before, while ETV6 has been already associated with FAMS. Sanger sequencing confirmed the mutation on the germline DNA in heterozygosis. The screening of 2 affected relatives still alive confirmed the presence of the variant in heterozygosis. In silico analysis performed on PolymiRST Database revealed that c.*514C>T in 3'UTR of ETV6 results in a gain of miRNA binding site: hsa-miR- 4717-3p and hsa-miR- 942-3p. Discussion The variant c.*514C>T in 3'UTR of ETV6 seems to repress ETV6 due to RNA interference. The new binding miRNAs have been already described as over-expressed in solid and hematologic tumors. Moreover, the down-regulation of ETV6 is associated with alteration of cell growth and hematopoiesis. Due to these evidences, c.*514C>T in 3'UTR of ETV6 could be considered as a new mutation involved in FAMS predisposition. Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
- Full Text
- View/download PDF
24. Parameters of Protein Metabolism and Thyroid Function As Predictors in a Scoring System for Acute and Chronic Graft-Versus-Host Disease
- Author
-
Michele Malagola, Valeria Cancelli, Andrea Di Palma, Carla Filì, Cristina Skert, Rosa Daffini, Simona Bernardi, Alessandro Turra, Chiara Pagani, Federica Cattina, Cesare Bergonzi, Rossella Ribolla, Domenico Russo, and Simone Perucca
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Immunology ,Protein metabolism ,Cell Biology ,Hematology ,Total body irradiation ,medicine.disease ,Biochemistry ,Thyroid function tests ,Gastroenterology ,Transplantation ,chemistry.chemical_compound ,Immune system ,Graft-versus-host disease ,chemistry ,Internal medicine ,medicine ,Liver function ,Thyroid function ,business - Abstract
Background Graft versus host disease (GVHD) is a common complication of allogeneic stem cell transplantation (allo-SCT), and represents its major cause of morbidity and mortality. Some “classical” patient-, donor- and transplant characteristics, such as age, gender disparity, donor type, HLA-match, and source of stem cells, have been reported as predictors for acute and chronic GVHD. However, no studies analysed these “classical” variables together with parameters of metabolic and endocrine functions, which may potentially influence the immune system. Thus, patient-and transplant variables together with index of liver and thyroid function, and some parameters of protein and lipid metabolism were retrospectively evaluated at different time points after transplantation, in order to identify possible predictors of acute and chronic GVHD and to calculate a risk score. Methods Clinical and transplant characteristics, number and type of infections before and after SCT were analysed in 161 patients. The following variables were also analysed pre-SCT, at day +7,+14,+21,+28, at + 3 and +6 months: LDH, parameters of liver function; parameters of protein and lipid metabolism (serum albumin, urea, total protein, cholesterol, triglycerides); thyroid function tests; autoimmune parameters (anti-nuclear, anti-DNA, anti-cardiolipin antibodies); body mass index. A 2-step multivariate analysis was performed using principal component analysis (PCA) and Cox regression analysis, in order to solve the problem of the high number of variables (metabolic, patients- and transplant related) in comparison with the relatively limited and heterogeneous pool of patients. Based on the regression coefficient of Cox analysis for each significant predictor, a scoring system for acute and chronic GVHD was calculated. Results In multivariate analysis, diagnosis of Myelodisplastic Syndrome or Chronic Myeloid Leukemia (HR 4,9; p=0.0004), conditioning regimen including Total Body Irradiation (HR 3,3; p=0.0003), and pre transplantation urea > 34 mg/dl with +21 day urea > 54 mg/dl (HR 2,6; p=0.0008) were evidenced as predictors for acute GVHD. Score values for each factor are 2, 1, and 1, respectively. Hence, the score, obtained by the sum of the three score values, ranged from 0 to 4. The probability of acute GVHD ranged from 8% (score 0) to 98% (score 4). Female donor (HR 5,1; p=0.0008), pre-SCT TSH values ≥ 2 mU/L with +28 day urea ≥ 39 mg/dl (HR 3,3; p=0.02), +6 month total protein < 5,5 g/dl with gamma-GT ≥ 347 U/L (HR 7,8, p=0.0001) resulted predictors for moderate/severe chronic GVHD, with score values of 1, 1, and 2 respectively. Risk of chronic GVHD at +6,5 month ranged from 3% (score 0) to 97% (score 4). Discussion Our study evidenced that factors other than “classical” ones may be associated to GVHD. Our scoring system includes routine-parameters (urea, total protein, gamma-GT,TSH), which are easily available in clinical practice. Urea levels depend on the balance between protein intake, endogenous catabolism and urinary excretion. The inflammatory microenvironment of GVHD promotes muscle catabolism, hence, urea levels do increase. Increased urea levels could be an indirect index of increased uremic toxins as well, which may stimulate the production of pro-inflammatory cytokines and the activation of leukocytes. On the other hand, increased urea levels and uremic toxins could derive from a dysregulated metabolism of the gut microbiome, which may influence immune system. Our findings, which require a prospective validation, suggest the usefulness a deeper study of the complex network between metabolic/endocrine functions and immune system, in order to develop a holistic approach of the transplant management. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
25. WT1 Monitoring of Minimal Residual Disease (MRD) in Patients with Acute Myeloid Leukemia
- Author
-
Crisitina Skert, Giuseppe Rossi, Giuseppina Ruggeri, Rossella Ribolla, Alessandro Turra, Valeria Cancelli, Enrico Morello, Francesca Antoniazzi, Michele Malagola, Domenico Russo, Andrea Di Palma, Elisa Alghisi, Simona Bernardi, Federica Cattina, Luigi Caimi, Simone Perucca, Chiara Pagani, and Erika Borlenghi
- Subjects
medicine.medical_specialty ,Univariate analysis ,ABL ,business.industry ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Gastroenterology ,Minimal residual disease ,Surgery ,Transplantation ,Leukemia ,medicine.anatomical_structure ,Internal medicine ,Medicine ,In patient ,Bone marrow ,business - Abstract
Background: Although a complete remission (CR) can be achieved in 70-80% of newly diagnosed acute myeloid leukemia (AML) patients, relapses occur in up to the 50% of cases. Thus, minimal residual disease (MRD) monitoring is a major issue for early detection of patients at high-risk of treatment failure and relapse. Aim: to dynamically evaluate WT1 pan-leukemic molecular marker of MRD in patients with AML. Matherial and methods: 107 newly diagnosed AML patients consecutively treated between 2010 and 2013 were monitored with quantitative WT-1 from bone marrow (BM) and peripheral blood (PB) at baseline, after induction, after the first consolidation course, before allogeneic stem cell transplantation (allo-SCT), at the 3rd and the 6th month after transplantation Results: At diagnosis, 104/107 (97%) had increased PB and BM WT1 levels assessed according to the ELN assay. Eighty-eight out of 107 patients (82%) achieved a complete remission (CR) after induction, 30/88 (34%) relapsed during follow up and 24/107 (22%) were addressed to allogeneic stem cell transplantation (allo-SCT). By univariate analysis, PB-WT > 50x10^4/ABL and BM-WT1 > 250x10^4/ABL after induction (PB: p=0.02; BM: p=0.04), after consolidation (PB: p=0.003), at the end of treatment (PB and BM: p=0.001), at 3rd month of follow up (PB and BM: p=0.005) and at 6th month of follow up (PB: p=0.005) were associated with a reduced overall survival (OS). By multivariate analysis, a BM-WT1 > 250 x 10^4/ABL at the end of treatment was significantly associated with a reduced OS. In order to adapt the cut-off of WT1 in our series of patients, we considered WT1 levels as continuous variables and categorized them at approximately the 25th, 50th, and 75th percentile. A cut-off of PB-WT1 > 25x10^4/ABL and BM-WT1 > 125x10^4/ABL at the end of the treatment program was identified as correlated with reduced leukemia-free survival (LFS) and OS (p=0.001). Similarly, and restricting the analysis on the 24 patients allo-transplanted in CR, 8/11 (73%) with pre-transplant PB-WT1 ≥ 5 and 4/13 (31%) with PB-WT1 < 5 relapsed, respectively (p=0.04). The incidence of relapse was higher in AML patients with PB-WT1 ≥ 5 measured at 3rd (56% vs 38%; p=0.43) and 6th month (71% vs 20%; p=0.03) after allo-SCT. Interestingly, 5/5 (100%) patients with pre-transplant PB-WT1 ≥ 5 who never reduced this level at 3rd or 6th month after allo-SCT experienced a disease recurrence. Conclusions: our data, although retrospectively collected, show that WT1 monitoring may be useful to predict the relapse in AML patients. Acknowledgments: This work was supported in part by Banca di Credito Cooperativo di Pompiano e Franciacorta and Lions Club Bassa Bresciana Association. Disclosures Russo: Celgene: Research Funding; Gilead: Research Funding; Novartis: Consultancy.
- Published
- 2014
26. SIRPB1 Is a Strong Predictor Biomarker of Response to 5-Azacitidine Therapy in MDS and AML Patients
- Author
-
Nicoletta Testoni, Simona Bernardi, Domenico Russo, Sarah Parisi, Carmen Baldazzi, Massimo Delledonne, Chiara Sartor, Francesca Volpato, Maria Chiara Fontana, Anna Maria Ferrari, Emanuela Ottaviani, Antonella Padella, Viviana Guadagnuolo, Michele Malagola, Carla Filì, Ilaria Iacobucci, Federica Cattina, Cristina Papayannidis, Stefania Paolini, Giovanni Martinelli, Maria Chiara Abbenante, Giorgia Simonetti, Guadagnuolo, Viviana, Papayannidis, Cristina, Iacobucci, Ilaria, Padella, Antonella, Simonetti, Giorgia, Paolini, Stefania, Abbenante, Mariachiara, Parisi, Sarah, Volpato, Francesca, Sartor, Chiara, Fontana, MARIA CHIARA, Ottaviani, Emanuela, Ferrari, Anna, Testoni, Nicoletta, Baldazzi, Carmen, Delledonne, Massimo, Fili', Carla, Malagola, Michele, Cattina, Federica, Bernardi, Simona, Russo, Domenico, and Martinelli, Giovanni
- Subjects
Oncology ,medicine.medical_specialty ,NPM1 ,Immunology ,Azacitidine ,Single-nucleotide polymorphism ,Cell Biology ,Hematology ,Biology ,Trisomy 8 ,medicine.disease ,Biochemistry ,Molecular biology ,IDH2 ,biomarcatore, 5-azacitidina ,ETV6 ,Internal medicine ,medicine ,Gene chip analysis ,ATRX ,medicine.drug - Abstract
Myelodisplastic syndromes (MDS) and Acute Myeloid Leukemia (AML) are a group of diseases of the elderly that initiates in a hematopoietic stem cell and are characterized by clonal hematopoiesis and uncertain prognosis, mostly due to cytogenetic background. In both diseases, 5-Azacitidine (5-Aza) has been successful, inducing prolonged survival and delayed AML evolution. To identify the genes mostly predictive of treatment response, we use high-throughput genomic analysis (SNP arrays and/or NGS-RNA-seq and/or NGS-WES and/or GEP) in azacitidine-sensitive and resistant MDS/AML patients. NGS-WES or RNA seq HiSeq 2000 (Illumina) was positively done in 35/214 AML samples (16%), GEP (GeneChip Human Transcriptome Array 2.0, Affymetrix Inc.) was performed in 65/214 AML samples (30%). SNPs arrays (CytoScan HD Array, Affymetrix Inc.) was done in 125/214 AML samples (58%) and 18/32 MDS samples (56%) at diagnosis, then analyzed by Chromosome Analysis Suite (ChAS) v1.2 (Affymetrix Inc.), Nexus Copy Number™ v7.5 (BioDiscovery) and GeneGo MetaCore™ software. We treated 246 adult patients (pts) with MDS or AML: 214 pts were AML and 32 were MDS with a median age of 59 and 70 years, respectively. Forty-five pts were treated with 5-Aza (32 MDS / 13 AML), while 201 AML were treated with conventional chemotherapy. Forty-five MDS/AML pts were treated with at least one complete cycle of 5-Aza (75 mg/sqm/daily). SNP arrays was done in 22/45 (49%), 13 pts were defined “insensitive/resistant”, ie. never achieving clinical complete remission (CCR) and 9 were defined “sensitive”, ie. all of them obtaining CCR. Copy Number Alterations (CNAs) ranged from loss or gain of complete chromosome (chr) arms to focal deletions and gains targeting one or few genes involving macroscopic (>1.5 Mbps), submicroscopic genomic intervals (50 Kbps - 1.5 Mbps) and LOH (>5 Mbps) events. Macroscopic CNAs affecting a complete chromosome or its arms were detected in 5 of 22 pts (23%), while classical cytogenetic was able to detect only two cases of trisomy 8 (9%), suggesting superiority of SNPs array for CNAs identifications. Microscopic CNAs abnormalities were detected in all of the patients affecting all the chromosomes. Of interest, some of them were located on chr 2, 3, 4, 5, 7, 8, 11, 12, 15, 17, 20, X, and were involved genes such as: NPM1, CTNNA1, IRF1, RPS14, SPARC, CBL, ETV6, EZH2, CUX1, CDC25C, EGR1, RUNX1, BRAF, ASXL1, ZRSR2, PHF6, BCOR, CDC25C, EGR1, IRAK1 in loss; RAD21, JAK2, KIT, ZRSR2, PHF6, BCOR, IRAK1 in gain; SIRPB1 both in loss and gain. Moreover we found in LOH these genes: NF1, GATA2, FADD, IDH2, SF3B1, BCOR, PHF6, ZRSR2, STAG2, KDM6A, ATRX, IRF1, NPM1, CBL, CTNNA1, EGR1, IRAK1. Chromosomic aberrations disease-related are more statistically frequent on pts “insensitive” versus pts. “sensitive” (64% vs. 35%) (p≤0.01). Moreover we found that from the median of chromosomic alterations lenghts (in kbp) the group of “insensitive” MDS/AML patients to 5-Aza therapy present more gains and losses than “sensitive” ones. By Nexus Copy Number software, we identify 137 genes highly differentially gain (SIRPB1 and KIT with p ≤ 0.05) or loss (SIRPB1, LCE1C, BCAS1, EXD3 with p ≤ 0.05) or LOH between “insensitive” versus “sensitive” to 5-Aza (p ≤ 0.05). Among these genes, we focused on SIRPB1 (cytoband 20p13, 56Kbps), since it was loss on 14/22 (64%) “insensitive” pts (p=0,023) and gain on 7/22 (70%) “sensitive” ones with a significantly (p=0,0324), respectively. SIRPB1 common deletion region goes from 1571 to 1598 (27 kbps) and common amplified region goes from 1561 to 1591 (30 kbps). By NGS-WES we analyzed 35/214 (16%) AML samples at diagnosis and we searched for point mutations, insertion/deletion or other abnormalities, involved in biomarkers of sensitivity/refractory to 5-Aza or chemoteraphy. We found mutations in SF3B1, NPM1, CBL, RUNX1, BCOR, KIT, GATA2, IDH2, KDM6A, KIAA1324L, PRIM2, RRN3, APOBR and again in SIRPB1 an heterozigosity frameshift deletion (c. 388delC; p. H130fs) in exone 2 in a AML pts with normal karyotype. By GEP we further analyzed 48/214 (22%) AML samples for SIRPB1 in order to correlate and confirm the expression or loss of expression of these genes, in correlation with 5-Aza response. We conclude that SIRPB1 is a promising marker of response to 5-Aza treatment in MDS and AML. Acknowledgments: ELN, AIL, AIRC, PRIN, progetto Regione-Università 2010-12 (L. Bolondi), FP7NGS-PTL project. Celgene ITA–VZ–MDS–PI-0298 GRANT-ITA-002 Disclosures Martinelli: Novartis: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau; Pfizer: Consultancy; ARIAD: Consultancy.
- Published
- 2014
27. Next-Generation Sequencing Analysis Revealed That BCL11B Chromosomal Translocation Cooperates with Point Mutations in the Pathogenesis of Acute Myeloid Leukemia
- Author
-
Francesca Volpato, Elisa Zago, Francesca Griggio, Emanuela Ottaviani, Massimo Delledonne, Giulia Paciello, Nicoletta Testoni, Antonella Padella, Viviana Guadagnuolo, Giovanni Martinelli, Simona Bernardi, Ilaria Iacobucci, Alberto Ferrarini, Anna Maria Ferrari, Elisa Ficarra, Carmen Baldazzi, Giorgia Simonetti, Maria Chiara Abbenante, Cristina Papayannidis, Marianna Garonzi, Raffaele A. Calogero, Padella, Antonella, Simonetti, Giorgia, Guadagnuolo, Viviana, Ottaviani, Emanuela, Ferrari, Anna, Zago, Elisa, Griggio, Francesca, Garonzi, Marianna, Paciello, Giulia, Bernardi, Simona, Baldazzi, Carmen, Papayannidis, Cristina, Abbenante, Mariachiara, Volpato, Francesca, Calogero, Raffaele, Testoni, Nicoletta, Ficarra, Elisa, Ferrarini, Alberto, Delledonne, Massimo, Iacobucci, Ilaria, and Martinelli, Giovanni
- Subjects
Genetics ,Sanger sequencing ,Myeloid ,BCL11B Gene ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Gene expression profiling ,Fusion gene ,symbols.namesake ,Leukemia ,medicine.anatomical_structure ,medicine ,symbols ,Next Generation Sequencing, Acute Myeloid Leukemia ,Exome - Abstract
Whole exome and transcriptome sequencing (WES and RNAseq) technologies are able to provide a comprehensive analysis of the genomic aberrations acquired by malignant cells, of their synergistic effects and functional consequences. In particular, RNAseq enables the detection of gene fusions originating from rare chromosomal translocations that have been involved in the pathogenesis of Acute Myeloid Leukemia (AML). We performed WES and RNAseq of AML patients to identify novel genetic abnormalities playing a causative role in leukemia development. We collected bone marrow or peripheral blood samples of 31 patients. Sequencing was performed using the Illumina Hiseq2000 platform. WES raw data were analysed with Whole-Exome sequencing Pipeline web tool for variants detection (WEP). The presence of gene fusions was assessed in RNAseq data with deFuse and Chimerascan. Selected genes fusions and variants were validated by Sanger sequencing. By RNAseq we identified a rare gene fusion transcript involving the BCL11B gene, which been previously suggested to play an oncogenic role in AML. The gene encodes for a zinc-finger protein participating to chromatin remodelling and regulating the differentiation and apoptosis of hematopoietic cells. The fusion was identified in a patient with poorly differentiated leukemia phenotype and unfavourable karyotypic abnormalities: 46,XX, t(2;14)(q21;q32), t(11;12)(p15;q22), who received standard chemotherapy, underwent allogeneic bone marrow transplantation and is currently in complete remission. Differently from previous data, the BCL11B translocation was associated neither with FLT3-ITD nor DNMT3A mutations. WES analysis revealed mutations in the TET2 and WTAP genes, which are known to act as co-players in the leukemic transformation. The exome data of our AML cohort identified neither INDELs nor nonsynonymous mutations in the BCL11Bgene, suggesting that the oncogenic function of BCL11B is activated by chromosomal translocations. Gene expression profiling showed a 4-fold upregulation of BCL11B transcript in the patient’s blasts, compared to 53 AML samples with no chromosomal aberrations in the 14q32 region, according to cytogenetic analysis. The increased expression of BCL11B was associated with an upregulation of potential targets including the antiapoptotic protein SPP1. Our data suggest that chromosomal translocations involving the BCL11B gene are rare events in AML and associate with somatic mutations in the malignant transformation of myeloid lineage cells, potentially by altering the differentiation and apoptotic processes. Future studies will investigate putative fusion partners of BCL11Band elucidate the biological consequences of its upregulation in AML pathogenesis. The results highlight the molecular heterogeneity of AML and the need for high-resolution sequencing analysis of leukemic samples at diagnosis in order to tailor personalized therapies. Supported by: FP7 NGS-PTL project, ELN, AIL, AIRC, PRIN, progetto Regione-Università 2010-12 (L. Bolondi). Disclosures Martinelli: Novartis: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau; Pfizer: Consultancy; ARIAD: Consultancy.
- Published
- 2014
28. A Gene Panel NGS-Based Strategy for Genomic Characterization of Acute Myeloid Leukemias (AMLs)
- Author
-
Michele Malagola, Francesca Schieppati, Flavio Mignone, Federica Cattina, Simona Bernardi, Crisitina Skert, Alessandro Turra, Mirko Farina, Chiara Cattaneo, Angela Passi, Erika Borlenghi, Simone Perucca, Giuseppe Rossi, Domenico Russo, Valeria Cancelli, Andrea Di Palma, and Carla Filì
- Subjects
Sanger sequencing ,Genetics ,Neuroblastoma RAS viral oncogene homolog ,NPM1 ,education.field_of_study ,Immunology ,Population ,Cell Biology ,Hematology ,Amplicon ,Gene mutation ,Biology ,Biochemistry ,DNA sequencing ,symbols.namesake ,hemic and lymphatic diseases ,Genomic Profile ,symbols ,education - Abstract
AMLs are clonal disorders characterized by high genomic heterogeneity and several chromosomal and molecular alterations affecting patients' outcome. In about 40% of AML patients who do not show any citogenetic alteration, sequencing analysis identified different gene mutations which play a pivotal role in leukemogenesis and have a negative prognostic impact: FLT3, ASXL1, TET2, IDH1, IDH2, RUNX1, CBL, CEBPα, DNMT3A and TP53. Conventional Sanger sequencing may detect clones representing more than 20% of the total tumor population, whereas Next Generation Sequencing (NGS) can identify mutations in less than 1% of leukemic cell burden. The detection of these variants is relevant because they can play an important role in driving drug resistance and disease relapse and for biologic risk assessment. To that purpose, we designed a 23-genes panel including: FLT3, DNMT3A, RUNX1, ASXL1, IDH1, IDH2, BCOR, NRAS, KRAS, TET2, TP53, U2AF1, ZRSR2, SF3B1, SRSF2, CBL, CEBPα, EZH2, NPM1, TERT, TERC, ETV6, GATA2. By using a 454 GS Junior by Roche Diagnostics with an amplicon-based sequencing approach and performing three sequencing runs per sample in order to reach a sensitivity of at least 1%, we settled an investigative strategy in order to assess the genomic profile of AMLs at diagnosis and possibly at the time of relapse or resistance. By this approach, we were able to confirm all the variants (i.e. FLT3, NPM1) previously documented with conventional tests; to reveal the presence of variants under the Sanger threshold of 20% in the genes resulted as wild type with routinely analysis; to evaluate the AML clonal heterogeneity, by assessing the coexistence of several mutated clones which have been described to be related to drug resistance, poor prognosis or to prior myelodysplastic syndrome. This gene panel NGS-based strategy may be proposed as a highly accurate and sensitive approach for genomic characterization of acute myeloid leukemias and as an useful tool for planning a target and personalized AML therapy. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
- Full Text
- View/download PDF
29. Index of Bone Marrow Output and Imbalance of B-Lymphocyte Homeostasis before and after Transplantation Correlate Differently with Graft-Versus-Host Disease and Relapse
- Author
-
Claudia Ghidini, Valeria Cancelli, Federica Cattina, Simone Perucca, Chiarini Marco, Simona Bernardi, Crisitina Skert, Viviana Giustini, Michele Malagola, Imberti Luisa, Carla Filì, Alessandro Turra, and Domenico Russo
- Subjects
T-cell receptor excision circles ,Lymphocyte ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Immune system ,Graft-versus-host disease ,medicine ,Bone marrow ,CD8 - Abstract
Introduction The long-term efficacy of allogeneic haematopoietic stem cell transplantation (SCT) relies primarily on the Graft-versus-tumor (GVT) effect, which partially overlaps with Graft versus Host disease (GvHD), the most common cause of morbidity and mortality in SCT. Researches on GVHD-biomarkers are still ongoing and a set of validate markers are still lacking, especially for chronic GVHD. Furthermore, immune parameters that univocally associate with GVHD or GVT have not been identified yet. In this study, lymphocyte subsets together with TCR-repertoire analysis, and index of thymic and bone marrow output were evaluated at different time points, in order to identify possible predictors of GVHD and ineffective GVT. Methods Prospective evaluations of lymphocyte subsets, thymic and bone marrow output were performed in 40 patients before SCT, at 30, 90, 180 days and 1 year after SCT. CD4+/CD8+ naïve, central memory, effector memory, terminally differentiated effector memory (TEMRA) cells, subsets of regulatory T-lymphocytes, immature B cells, naïve, switched and unswitched memory B cells, memory double negative (IgD-CD27-) B cells were analysed by flow cytometry. Analysis of thymic and bone marrow output was performed by detection of T cell receptor excision circles (TRECs) and kappa-deleting recombination circles (KRECs). TRECs and KRECs were simultaneously quantified by a duplex quantitative Real-Time PCR. Heteroduplex assay was used to perform TCR-repertoire analysis. A 2-step multivariate analysis was performed using principal component analysis (PCA) and Cox regression analysis, to solve the problem of the high number of variables (immunological, patients- and transplant related) in comparison with the relatively limited and heterogeneous pool of patients. Results Twenty patients developed acute GVHD (median time: 28 days, range 19-120). Chronic GVHD was observed in 9 patients (median time: 6 months, range 4-10). In multivariate analysis, acute GVHD correlated positively with pre-transplant percentage of CD4+ central memory cells, and with values of regulatory effector memory T-cells and CD4+TEMRA cell at day +30 (p=0,0006). Pre-transplant percentage of unswitched memory B cells was also associated with acute GVHD, whereas pre-transplant levels of KRECs were inversely correlated (p=0,0005). Chronic GVHD was associated with matched unrelated donor and with (p -values of regulatory effector memory T-cells at +30, percentage of CD8+TEMRA cells at +90, values of immature B cells and levels of KRECs at +180 (positive correlation) -percentage of CD4+ central memory and CD8+ effector memory cells at +90 (negative correlation). The relapse rate (27%; median time: 5,5 months, range 3-12) was used as clinical index of ineffective GVT. The following cluster of immunological parameters at day +90 correlated positively with relapse: CD8+ effector memory cells, immature B cells, naïve, switched memory B cells, memory double negative (IgD-CD27-) B cells (p=0,006). Discussion Different clusters of immunological parameters at different time points were evidenced as predictors of GVHD and ineffective GVT, allowing a clear-cut distinction between these immunological reactions. Changes in pre- and post-transplant B-lymphopoietic microenvironment and specific imbalances in the subset of B-cells may be involved in acute and chronic GVHD development. The atypical association of regulatory T-cells with GVHD may be explained by the relative efficiency of different subsets of regulatory T-cells (naïve>effector memory), as shown in some experimental models. Increased values of CD8+ effector memory cells could be an early sign of ineffective GVL. Imbalance toward a lymphocyte B-response, and especially toward "senescent" memory (IgD-CD27-) B cells, could promote tolerance to tumor cells. The validation of these clusters of immunological parameters as specific early predictors of GVHD or GVT, even before SCT, could potentially allow the development of pre-emptive and targeted therapies. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
- Full Text
- View/download PDF
30. A Specific Pattern of Somatic Mutations Associates with Poor Prognosis Aneuploid Acute Myeloid Leukemia: Results from the European NGS-PTL Consortium
- Author
-
Ilaria Iacobucci, Antonella Padella, Massimo Delledonne, Maria Chiara Abbenante, Emanuela Ottaviani, Giovanni Martinelli, Nicoletta Testoni, Daniel Remondini, Simona Bernardi, Marco Manfrini, Viviana Guadagnuolo, Alberto Ferrarini, Giovanni Marconi, Annalisa Astolfi, Carmen Baldazzi, Simona Soverini, Torsten Haferlach, Michele Cavo, Elisa Zago, Cristina Papayannidis, Marianna Garonzi, Giorgia Simonetti, Italo Faria do Valle, Anna Maria Ferrari, Maria Chiara Fontana, Simonetti, Giorgia, Padella, Antonella, FARIA DO VALLE, Italo, Manfrini, Marco, Papayannidis, Cristina, Baldazzi, Carmen, Fontana, MARIA CHIARA, Guadagnuolo, Viviana, Ferrari, Anna, Zago, Elisa, Garonzi, Marianna, Bernardi, Simona, Ottaviani, Emanuela, Astolfi, Annalisa, Abbenante, Mariachiara, Marconi, Giovanni, Soverini, Simona, Cavo, Michele, Testoni, Nicoletta, Ferrarini, Alberto, Delledonne, Massimo, Haferlach, Torsten, Remondini, Daniel, Iacobucci, Ilaria, and Martinelli, Giovanni
- Subjects
Genome instability ,Genetics ,Monosomy ,Mutation ,Immunology ,Aneuploidy ,Chromosome ,Cell Biology ,Hematology ,Cell cycle ,Biology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Leukemia ,Acute Myeloid Leukemia, Aneuploidy ,hemic and lymphatic diseases ,Chromosome instability ,medicine ,neoplasms - Abstract
Aneuploidy causes a proliferative disadvantage, mitotic and proteotoxic stress in non-malignant cells ( Torres et al. Science 2007). Chromosome gain or loss, which is the hallmark of aneuploidy, is a relatively common event in Acute Myeloid Leukemia (AML). About 10% of adult AML display isolated trisomy 8, 11, 13, 21 (Farag et al. IJO 2002), or either an isolated autosomal monosomy or monosomal karyotype (Breems et al. JCO 2008). This evidence suggests that tumor-specific mechanisms cooperate to overcome the unfitness barrier and maintain aneuploidy. However, the molecular bases of aneuploid AML are incompletely understood. We analyzed a cohort of 166 cytogenetically-characterized AML patients (80 aneuploid (A-) and 86 euploid (E-)) treated at Seràgnoli Institute (Bologna). Aneuploidy was significantly associated with poor overall survival (median survival: 13 and 26 months in A-AML and E-AML respectively; p=.006, Fig.1). To identify AML-specific alterations having a causative and/or tolerogenic role towards aneuploidy, we integrated high-throughput genomic and transcriptomic analyses. We performed 100 bp paired-end whole exome sequencing (WES, Illumina Hiseq2000) of 70 samples from our A-AML and E-AML cohort of 166 patients. Variants where called with MuTect or GATK for single nucleotide variant and indels detection, respectively. AML samples were genotyped by CytoScan HD Array (Affymetrix). Gene expression profiling (GEP) was also conducted on bone marrow cells from 24 A-AML, 33 E-AML (≥80% blasts) and 7 healthy controls (HTA 2.0, Affymetrix). We detected a significantly higher mutation load in A-AML compared with E-AML (median number of variants: 31 and 15, p=.04) which was interestingly unrelated to patients' age (median age: 63.5 years in A-AML and 62 years in E-AML, Xie et al, Nat. Med. 2014). C>A and C>T substitutions, which are likely mediated by endogenous 5mCdeamination, were the most frequent alterations (Alexandrov et al. Nat. 2013). However, aneuploidy associated with an increased variability in terms of mutational signatures, with the majority of A-AML displaying 3 or more signatures compared to few E-AML cases (p=.04). WES analysis also revealed a specific pattern of somatic mutations in A-AML. A-AML had a lower number of mutations in signaling genes (p=.04), while being enriched for alterations in cell cycle genes (p=.01) compared with E-AML. The mutated genes were involved in different cell cycle phases, including DNA replication (MCM6, PURB, SSRP1), centrosome dynamics (CEP250, SAC3D1, HEPACAM2, CCP110), chromosome segregation (NUSAP1, ESPL1, TRIOBP), mitotic checkpoint (ANAPC7, FAM64A) and regulation (CDK9, MELK, ZBTB17, FOXN3, PPM1D, USP2). Moreover, genomic deletion of cell cycle-related genes was frequently detected in A-AML. Notably, ESPL1 which associated with aneuploidy, chromosome instability and DNA damage in mammary tumors (Mukherjee et al. Oncogene 2014) was mutated and also upregulated in A-AML compared with E-AML (p=.01), the latter showing expression levels comparable to controls. Among the top-ranked genes differentially expressed between A-AML and E-AML, we identified a specific signature characterized by increased CDC20 and UBE2C and reduced RAD50 and ATR in A-AML (p Our data show a link between aneuploidy and genomic instability in AML. Deregulation of the cell cycle machinery, DNA damage and repair checkpoints either through mutations, copy number and transcriptomic alterations is a hallmark of A-AML. The results define specific genomic and transcriptomic signatures that cooperate with leukemogenic pathways, as KRAS signaling, to the development of the aggressive phenotype of A-AML and suggest that a number of A-AML patients may benefit frompharmacological reactivation of TP53pathway (e.g. MDM2 inhibitor, clinical trial NP28679). Supported by: FP7 NGS-PTL project, ELN, AIL, AIRC, PRIN, progetto Regione-Università 2010-12 GS & AP: equal contribution Disclosures Soverini: Novartis, Briston-Myers Squibb, ARIAD: Consultancy. Cavo:JANSSEN, CELGENE, AMGEN: Consultancy. Haferlach:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Martinelli:MSD: Consultancy; BMS: Speakers Bureau; Roche: Consultancy; ARIAD: Consultancy; Novartis: Speakers Bureau; Pfizer: Consultancy.
- Published
- 2015
- Full Text
- View/download PDF
31. Genomic Analisys of Notch Mutations in a Case of Alagille Syndrome with Acute Lymphoblastic Leukemia
- Author
-
Giulio Bassi, Omar Perbellini, Paul Takam Kamga, Massimo Delledonne, Mauro Krampera, Achille Ambrosetti, Giovanni Martinelli, Simona Bernardi, Luigi Scaffidi, Giovanni Pizzolo, Sergio Marin Vargas, Ilaria Iacobucci, Paola Tononi, and V. Meneghini
- Subjects
JAG1 ,Pathology ,medicine.medical_specialty ,Immunology ,Autosomal dominant trait ,Germline mosaicism ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Penetrance ,Alagille syndrome ,medicine ,Jagged-1 Protein ,Neoplastic transformation ,Exome - Abstract
Alagille syndrome (ALGS), or arteriohepatic dysplasia, is a congenital multisystem disease due to Notch signalling pathway mutations, most commonly affecting JAGGED-1 (ALGS type 1), and more rarely NOTCH-2 (ALGS type 2), leading to hepatic, lung, renal and ocular dysfunction (chronic cholestasis, peripheral pulmonary artery stenosis, dysplastic kidneys pigmentary retinopathy), and skeletal abnormalies (minor vertebral segmentation, characteristic facies, posterior embryotoxon/anterior segment defects). ALGS is an autosomal dominant disease, but it is characterized also by variable penetrance and clinical expression and somatic/germline mosaicism. A 20-year-old man with ALGS was admitted to the University Hospital of Verona because of pancytopenia. Following analyses led to the diagnosis of Philadelphia chromosome/bcr-abl-negative, CD10-positive, B-lineage acute lymphoblastic leukemia (common B-ALL). In order to identify the genetic components involved in this complex phenotype, we sequenced the exome of a bone marrow sample collected from the patient. By genome interpretation with Knome pipeline applied to the reference genome UCSC hg19, we found missense variants both in NOTCH-2 (E38K) and JAGGED-1 (P871R) genes that are mainly involved in the syndrome, although their effect on protein function was predicted not to be deleterious. However, we detected putative damaging mutations in genes such as PAX5 (R38H) and NOTCH-1 (K1821N), which might be strongly related to the observed disease. In fact, PAX5 is a member of PAX protein family of transcription factors implicated into regulation of early development that binds NOTCH-2 and likely altering its functionality. On the other hand, NOTCH-1 is involved in cell growth and proliferation and thus the predicted alteration of function of the corresponding protein may have an important role in neoplastic transformation. Disclosures Martinelli: Novartis: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau; Pfizer: Consultancy; ARIAD: Consultancy.
- Published
- 2014
- Full Text
- View/download PDF
32. Genomic Analysis Of Notch Mutations In a Case Of Alagille Syndrome With Acute Lymphoblastic Leukemia
- Author
-
Simona Bernardi, Paola Tononi, Sergio Marin Vargas, Giulio Bassi, Achille Ambrosetti, Vittorio Meneghini, Luigi Scaffidi, Giovanni Martinelli, Ilaria Iacobucci, Giovanni Pizzolo, Massimo Delledonne, and Mauro Krampera
- Subjects
hemic and lymphatic diseases ,Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Alagille syndrome (ALGS), or arteriohepatic dysplasia, is a congenital multisystem disease due to Notch signalling pathway mutations, most commonly affecting JAG1 (ALGS type 1), and more rarely NOTCH2 (ALGS type 2), leading to hepatic, lung, renal and ocular dysfunction (chronic cholestasis, peripheral pulmonary artery stenosis, dysplastic kidneys pigmentary retinopathy), and skeletal abnormalies (minor vertebral segmentation, characteristic facies, posterior embryotoxon/anterior segment defects). ALGS is an autosomal dominant disease, but it is characterized also by variable penetrance and clinical expression and somatic/germline mosaicism. A 20-year-old man with ALGS was admitted to the University Hospital of Verona because of pancytopenia. Following analyses led to the diagnosis of Philadelphia chromosome/bcr-abl-negative, CD10-positive, B-lineage acute lymphoblastic leukemia (common B-ALL). In order to identify the genetic components involved in this complex phenotype, we sequenced the exome of a bone marrow sample collected from the patient. By genome interpretation with Knome pipeline applied to the reference genome UCSC hg19, we found missense variants both in NOTCH2 (E38K) and JAG1 (P871R) genes that are mainly involved in the syndrome, although their effect on protein function was predicted not to be deleterious. However, we detected putative damaging mutations in genes such as PAX5 (R38H) and NOTCH1 (K1821N) which might be strongly related to the observed disease. In fact, PAX5 is a member of PAX protein family of transcription factors implicated into regulation of early development, that binds NOTCH2 and likely altering its functionality. On the other hand, NOTCH1 is involved in cell growth and proliferation and thus the predicted alteration of function of the corresponding protein may have an important role in neoplastic transformation. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
- Full Text
- View/download PDF
33. Targeting HRASV12G Expression to the Zebrafish Early Hemogenic Progenitors Induces a Myeloproliferative Disorder by Repressing the Notch Pathway
- Author
-
Alessandro Turra, Carla Filì, Christiaan V. Henkel, Elisa Alghisi, Martin Distel, Michele Malagola, Cristina Skert, Cristina Santoriello, Rossella Ribolla, Valeria Cancelli, Domenico Russo, Marina Mione, Andrea Di Palma, Simona Bernardi, Cesare Bergonzi, Federica Cattina, Simona Zedda, and Simone Perucca
- Subjects
LMO2 ,biology ,Immunology ,Hematopoietic Tissue ,Notch signaling pathway ,Cell Biology ,Hematology ,biology.organism_classification ,Biochemistry ,Cell biology ,Haematopoiesis ,Myeloproliferative Disorders ,Erythrocyte differentiation ,Neoplastic transformation ,Zebrafish - Abstract
Abstract 4676 Introduction Myeloproliferative diseases (MPDs) are a group of haematological disorders characterized by the hyper proliferation of different blood cells in peripheral blood and other hematopoietic organs. The clinical heterogenity of these neoplasms reflects the different gene pathways involved, most of which are only partially known. Given the genetic homology and the physiological similarity to mammals, zebrafish has emerged as an ideal model to study human normal and malignant haematopoiesis. In the last decade several oncogenes involved in the development of hematopoietic neoplasms have been used to model leukemia in zebrafish with the aim to discovery new molecular pathways involved in malignant transformation. Despite the first encouraging results these experimental models failed to fully recapitulate human myeloproliferative disorders. Methods We took advantage of the Gal4/UAS binary system to induce the expression of human oncogenic HRASV12G in the zebrafish hematopoietic compartment. We used a specific transgenic line that drives oncogene expression in zebrafish early hematopoietic progenitors under control of the FLI.1 (Friend Leukemia virus Integration 1) promoter. Results We observed the development of a myelo-erythroid proliferative disease in few days in zebrafish transgenic larva. The pathological phenotype is characterized by the expansion of the hematopoietic tissue, an increased expression of myelo-erythroid specific genes (PU.1, gata1, mpx, c-mpl) associated with a slight increase of staminality markers (lmo2, scl, c-myb, runx.1), and a higher number of l-plastin expressing cells. Moreover blood smear of pathological larva displayed leukemic blasts and the arrest of erythrocyte differentiation whereas kidney marrow of juvenile fish displayed abnormal myelopoiesis characterized by the increase of erythro-myeloid progenitors. We found that the pathological phenotype is associated with a down regulation of the Notch pathway as shown by the decreased gene expression of notch pathways target genes (notch1, notch3, her6). Furthermore we discovered a novel set of genes involved in neoplastic transformation induced by HRASV12 expression through RNA-Seq analysis of pathological larva. Conclusions The expansion of the zebrafish hematopoietic compartment characterized by the hyper-proliferation of the myelo-erythroid progenitors that we found in this model reproduces some of the pathological features of human myeloproliferative disorders. This study showed that forcing oncogene expression in the hemogenic endothelial cells induces the transdifferentiation of the early hemogenic pluripotent stem cells into abnormal myeloerythoid progenitors by repressing the Notch pathways. Transcriptome analysis identified a number of potential effectors of this transformation. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.