81 results on '"Marco Ladetto"'
Search Results
2. Impact of COVID-19 Pandemic Waves on Outcomes of Patients with Previously Untreated Advanced Follicular Lymphoma Enrolled in the Urban Ambispective Study in Italy
- Author
-
Antonio Pinto, Emanuele Guardalben, Marica Battista, Giulia Chiara Gazzoli, Michele Merli, Annalisa Chiarenza, Tommasina Perrone, Attilio Guarini, Nicola Di Renzo, Carlo Visco, Agostino Tafuri, Roberta Murru, Felicetto Ferrara, Jacopo Olivieri, Attilio Olivieri, Andrés J M Ferreri, Marco Ladetto, Pier Luigi Zinzani, Luca Arcaini, and Giuseppe Gritti
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
3. Six-Month Doxycycline Is Safe and Effective As Upfront Monotherapy for Stage-I Malt Lymphoma of the Ocular Adnexae: Primary Endpoint Results of the IELSG39 Trial
- Author
-
Andrés J.M. Ferreri, Marianna C. Sassone, Maria Giulia Cangi, Gilda Magliacane, Stefania Zanussi, Elena Flospergher, Fabrizio Marino, Lucia Bongiovanni, Marco Ladetto, Federica Cavallo, Franco Aversa, Antonella Anastasia, Donato Mannina, Anna Pascarella, Daniele Vallisa, Alessandro Pulsoni, Luisella Bonomini, Francesco Bertoni, Riccardo Dolcetti, Emanuele Zucca, and Maurilio Ponzoni
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
4. Efficacy and Safety of Ibrutinib Combined with Standard First-Line Treatment or As Substitute for Autologous Stem Cell Transplantation in Younger Patients with Mantle Cell Lymphoma: Results from the Randomized Triangle Trial By the European MCL Network
- Author
-
Martin Dreyling, Jeanette K. Doorduijn, Eva Gine, Mats Jerkeman, Jan Walewski, Martin Hutchings, Ulrich Mey, Jon Riise, Marek Trneny, Vibeke K.J. Vergote, Melania Celli, Ofer Shpilberg, Maria Gomes da Silva, Sirpa Leppa, Linmiao Jiang, Christiane Pott, Wolfram Klapper, Döndü Gözel, Christian Schmidt, Michael Unterhalt, Marco Ladetto, and Eva Hoster
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
5. Minimal Residual Disease-Driven Treatment Intensification By Sequential Addition of Ibrutinib to Venetoclax in Relapsed/Refractory Chronic Lymphocytic Leukemia: Results of the Monotherapy and Combination Phases of the Improve Study
- Author
-
Pamela Ranghetti, Elisa Albi, Maria Colia, Eloise Scarano, Paolo Ghia, Andrea Ferrario, Marco Ladetto, Antonella Capasso, Silvia Heltai, Luana Schiattone, Rosaria Sancetta, Luca Laurenti, Marzia Varettoni, Lydia Scarfò, Marina Deodato, Eleonora Perotta, Gianluca Gaidano, Gianluigi Reda, Marina Motta, Marta Coscia, and Lucia Farina
- Subjects
medicine.medical_specialty ,business.industry ,Venetoclax ,Treatment intensification ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Minimal residual disease ,Discontinuation ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Ibrutinib ,Relapsed refractory ,Ven ,Clinical endpoint ,Medicine ,business - Abstract
The treatment of chronic lymphocytic leukemia (CLL) has been radically changed in the last years thanks to the targeted therapies, including kinase (i.e. ibrutinib) and BCL2 (i.e. venetoclax) inhibitors. Venetoclax (VEN) in particular is able to obtain undetectable minimal residual disease (uMRD), though only in a proportion of patients (pts) when given as single agent, thus warranting the need of different strategies in those not achieving uMRD. We designed a phase 2 multicenter Italian study where ibrutinib (IBR) is added to VEN based on a MRD-driven strategy aiming at obtaining uMRD and discontinuing both treatments in pts who did not achieve uMRD with VEN mono. Study treatment started with VEN (ramp up to 400 mg/day as per current label) for 12 months. MRD status in peripheral blood (PB) and bone marrow (BM) was evaluated using the 6-color flow cytometry assay recommended by ERIC (CD5/CD81/CD79b/CD19/CD43/CD20). Pts with uMRD in both PB and BM at C12D1 discontinued VEN at C12D28 and entered the follow-up phase. Pts with detectable MRD in PB and/or BM added IBR 420 mg/day starting from C13D1 and continued both drugs up to maximum C24D28, uMRD, progression or unacceptable toxicity (whichever occurs first). After C24D28, pts with detectable MRD and still in response continued IBR alone. The primary endpoint was uMRD4 ( Thirty-eight pts (recruited from Nov 2017 to Jul 2018) fulfilled eligibility and started VEN. Baseline characteristics included: median number of prior therapies 1 (range 1-4) (60.6% previously treated with FCR or FC); del(17p) in 8/33 (24%); TP53 mutations in 10/30 (33%), and unmutated IGHV in 24/30 (80%). At the data cut-off, 35/38 evaluable pts still in the study have reached C24D1, 1 pt discontinued treatment due to myelodisplasia (considered unrelated to study treatment) before C12D1 and 1 pt progressed on VEN monotherapy shortly before that timepoint, 1 evaluation is still missing due to COVID-19 restrictions. At C12D1, uMRD4 in PB was achieved in 19/38 (50%) pts (Figure 1), 17/19 (89.5%) had uMRD4 confirmed in BM. Overall response rate with VEN single-agent was 36/38 (94.7%), 9 CR and 27 PR. As per protocol, the 17 pts (45%) with uMRD4 in PB and BM at C12D1 discontinued VEN at C12D28. Nineteen responsive cases with detectable MRD at C12D1 added IBR to VEN starting from C13D1. The combination of IBR and VEN led to an improved reduction of the depth of MRD in all but 3 pts with 16/19 (84%) achieving uMRD4 in both PB and BM between C16D1 (first MRD assessment after starting IBR) and C24D1, thus stopping both therapies as per protocol. After a median follow-up of 25.4 months (range 6.1-33.5) from treatment initiation, no clinical progression was observed among those discontinuing treatment in uMRD, while MRD4 relapse occurred in 21/33. Median time to MRD4 relapse in those who achieved uMRD at any timepoint and discontinued treatment was 4 months (range 2-13). Twelve pts (6 treated with VEN only) remain uMRD after stopping treatment, with a median observation of 13 months (range 3+-18+) since confirmed uMRD4. Safety data were analyzed in the intention-to-treat cohort (39 pts). No cases of clinical tumor lysis syndrome (TLS) and/or biochemical TLS were reported in the 39 pts exposed to VEN. Adverse events (AEs) were mild, with no treatment discontinuations or dose reductions. Five Serious AEs (Table 1) and 130 AEs (Table 2) occurred in 28 patients, without any SUSARs. All 5 SAEs were deemed unrelated to study drug(s) and 4/5 have resolved without sequelae. In conclusion, we here present the updated results of our study including the combination phase of VEN with IBR. This sequential MRD-guided approach was feasible and led to deeper responses in about 85% of pts not achieving uMRD4 after VEN alone. With this tailored and time-limited strategy 33 out of 38 pts (87%) obtained uMRD4 in PB and BM either after VEN monotherapy or the IBR-VEN combination, indicating we may reach identical depth of response with a personalized intensification and avoid unnecessary drug exposure. Time to clinical progression and response to VEN retreatment in this cohort remain to be established as well as the biological characteristics of those pts with persistent MRD despite the combined treatment. Updated results with further sequential MRD and clinical monitoring after treatment discontinuation will be presented at the meeting. Disclosures Scarfo: Gilead: Membership on an entity's Board of Directors or advisory committees; AstraZeneca: Honoraria; Abbvie: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Farina:Abbvie: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Gaidano:Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Abbvie: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Astrazeneca: Membership on an entity's Board of Directors or advisory committees; Sunesys: Membership on an entity's Board of Directors or advisory committees. Reda:Janssen: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees. Coscia:Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees; Shire: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Karyopharm Therapeutics: Research Funding. Laurenti:Roche: Honoraria; Gilead: Honoraria; Janssen: Honoraria; AbbVie: Honoraria. Varettoni:Roche: Consultancy, Membership on an entity's Board of Directors or advisory committees; AbbVie: Other: Travel/accommodations/expenses; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Travel/accommodations/expenses. Ghia:Lilly: Consultancy, Honoraria; Sunesis: Consultancy, Honoraria, Research Funding; Adaptive, Dynamo: Consultancy, Honoraria; MEI: Consultancy, Honoraria; Celgene/Juno: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company), Research Funding; BeiGene: Consultancy, Honoraria; Acerta/AstraZeneca: Consultancy, Honoraria; ArQule: Consultancy, Honoraria; Gilead: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company), Research Funding; Novartis: Research Funding.
- Published
- 2020
6. Pharmacogenomics Drives Lenalidomide Efficacy and MRD Kinetics in Mantle Cell Lymphoma after Autologous Transplantation: Results from the MCL0208 Multicenter, Phase III, Randomized Clinical Trial from the Fondazione Italiana Linfomi (FIL)
- Author
-
Alice Di Rocco, Gomes da Silva Maria, Monica Balzarotti, Gian Maria Zaccaria, Giuseppe A. Palumbo, Anna Lia Molinari, Elisa Genuardi, Filippo Ballerini, Carola Boccomini, Marco Ladetto, Simone Ferrero, Beatrice Alessandria, Sergio Cortelazzo, Alessandro Re, Daniele Grimaldi, Vittorio Ruggero Zilioli, Federica Cavallo, Andrés J.M. Ferreri, Vittorio Stefoni, Luca Arcaini, Antonello Di Paolo, Elena Arrigoni, Marco Ghislieri, Benedetta Puccini, Sara Galimberti, and Gabriele De Luca
- Subjects
Oncology ,medicine.medical_specialty ,Haploview ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Minimal residual disease ,law.invention ,Efficacy ,Randomized controlled trial ,law ,Internal medicine ,Pharmacogenomics ,medicine ,Autologous transplantation ,Mantle cell lymphoma ,business ,Lenalidomide ,medicine.drug - Abstract
Background and Aims. Prediction of treatment efficacy is an active and growing field of pharmacology. In the Fondazione Italiana Linfomi (FIL) MCL0208 phase III trial (NCT02354313), a 24 months lenalidomide maintenance (LM, 15 mg days 1-21 every 28 days) after high-dose immuno-chemotherapy followed by autologous transplantation (ASCT) in 300 frontline mantle cell lymphoma (MCL) patients showed substantial clinical activity in terms of Progression-Free Survival (PFS) vs observation (OBS). However, this benefit seemed not uniform across patient series. To deeper investigate the differential pattern of response to lenalidomide, a wide analysis of the host pharmacogenomics (PG) background was planned, in order to dissect whether specific germline polymorphisms of transmembrane transporters, metabolic enzymes or cell surface receptors (ABCB1, ABCG2, VEGFA, FCGR2A, NCF4, GSTP1, CRBN) might predict the drug efficacy. Actually, several single nucleotide polymorphisms (SNPs) of ABCB1 exert an effect on substrate affinity of lenalidomide for the transmembrane transporter. Moreover, VEGFA is involved in the anti-angiogenic activity of lenalidomide and might eventually upregulate ABCB1 expression, too. Patients and methods. Genotypes for SNPs were obtained through allele-specific (ASO) probes on germline DNA from peripheral blood. Minor allele frequencies (MAFs) were obtained and the Hardy-Weinberg equilibrium (HWE) was checked. Genotypes were used to infer individual haplotypes by Arlequin and Haploview softwares. Minimal residual disease (MRD) was assessed with ASO primers on either IGH or BCL-1/IGH rearrangements by RQ-PCR in bone marrow samples. TP53 disruption was identified by NGS targeting resequencing and copy number variation analysis. Clinical-biological correlations were screened by automated machine learning methods and validated by both Kaplan-Meier at univariate level and Cox models for multivariate analysis (MV). A logistic regression was implemented to investigate correlations between polymorphisms and MRD kinetics. Results. 278 out of 300 patients (93%) were fully genotyped. The MAF values of the SNPs were very similar to published data and the HWE was confirmed. Most notably, ABCB1 c.2677G>T/A(W) and VEGFA c.2055A>C were significantly associated to outcome and are thus described in this abstract. In the case of ABCB1, the three loci were in strong linkage disequilibrium (p Disclosures Ferrero: Servier: Speakers Bureau; Gilead: Research Funding, Speakers Bureau; EUSA Pharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Boccomini:SC Ematologia, ASOU Città della Salute e della Scienza di Torino, Turin, Italy: Current Employment. Maria:Roche: Consultancy, Other: travel, accomodations, expenses; Abbvie: Consultancy, Other: travel, accomodations, expenses; BMS: Consultancy; MSD: Consultancy; Janssen: Consultancy, Other: travel, accomodations, expenses; Gilead: Consultancy, Other: travel, accomodations, expenses, Research Funding. Ferreri:Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; Morphosys: Research Funding; Hutchinson: Research Funding; BMS: Research Funding. Palumbo:Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Galimberti:Novartis: Speakers Bureau; Incyte: Honoraria. OffLabel Disclosure: Lenalidomide maintenance in mantle cell lymphoma
- Published
- 2020
7. Daratumumab after Allogeneic Hematopoietic Stem Cell Transplantation in Multiple Myeloma: Safety and Efficacy. a Retrospective Study from the Cmwp EBMT
- Author
-
Raphael Teipel, Marco Ladetto, Patrick Hayden, Laure Vincent, Linda Koster, Mathias Haenel, Luuk Gras, Marie Robin, Monique C. Minnema, Wilfried Schroyens, Jaime Sanz, Tsila Zuckerman, Meral Beksac, Liesbeth C. de Wreede, Sophie Ducastelle, Jürgen Finke, Edouard Forcade, JA Van Deosum, Christian Koenecke, Pascal Lenain, Ibrahim Yakoub-Agha, Laura Rosiñol, Patrice Ceballos, Jakob Passweg, and Stefan Schönland
- Subjects
Plasma cell leukemia ,medicine.medical_specialty ,business.industry ,Incidence (epidemiology) ,medicine.medical_treatment ,Immunology ,Daratumumab ,Retrospective cohort study ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Dara ,medicine.disease ,Biochemistry ,Transplantation ,Internal medicine ,medicine ,business ,Multiple myeloma - Abstract
BACKGROUND Allogeneic stem cell transplantation (allo-HSCT) is a treatment option for high-risk multiple myeloma (MM), especially in patients who relapse early following auto-HSCT. Though there is a proven graft versus myeloma effect, relapse remains common. Daratumumab (Dara) is a humanized monoclonal anti-CD38 antibody approved for both newly diagnosed and relapsed MM. Its mechanisms of action include direct anti-MM activity (CDC, ADCC, ADCP, apoptosis induction) and indirect anti-MM activity depleting CD38+ immunosuppressive regulatory cellsand promoting T-cell expansion and activation. The combination of its mechanism of action and lack of toxicity makes Dara a good candidate for use in the post-allo-HSCT setting. However, its immune effects (decrease in CD38-positive immune suppressor cells, including Tregs, NK cells, regulatory B cells, and myeloid-derived suppressor cells) may interfere with post-allo anti-MM effects. Nikolaenko et al (Clin Lymph Myeloma Leuk 2020) reported that aGVHD developed in five (15%) of 34 patients given Dara (mostly in combination) as treatment for post-allo relapse and the median PFS was 4.5 months. METHODS We performed a retrospective study to evaluate the safety and efficacy of Dara post-allo-HSCT). Patients with MM having received at least one Dara infusion at any time after allo-HSCT were included. Key exclusion criteria were plasma cell leukemia and AL amyloidosis. RESULTS A total of 121 patients who received Dara after a first allo-HSCT were identified in the EBMT database. The year of allo-HSCT ranged from 2004 to 2019, median 2014. Allo-HSCT was performed at a median (range) of 34 (6-172) months after the diagnosis of myeloma. The stem cell source was PB in 89%, 37% were matched related donor and 39% matched unrelated donor. Conditioning was reduced intensity in 72% and myeloablative in 28%. Disease status at allo-HSCT was CR in 9%, VGPR in 35 %, PR in 43%, SD/MR in 7% and progression in 6%. The median age (range) at the first Dara infusion was 55 (32-71) yrs with a male to female distribution of 70/51. Dara was administered either alone (n=70) or in combination with other anti-myeloma directed therapy (n=51). The first dose of Dara was given at a median (range) of 30 (1-173) months post-allo-HSCT. Fifteen patients started Dara in the first 6 months after allo-HSCT, 50% of patients in the first 2.5 years, 22% in 2.5 to 5 years, and 28% more than five years after allo-HSCT. Among patients with available data, 45% had at least one serious infection: bacteremia 22% (including 15% ³ grade 3), septic shock 5% (all ³ grade 3), pneumonia 31% (including 21% ³ grade 3), urine infections 7%, CMV reactivation 7% and EBV reactivation 6%. In the first 100 days after starting Dara, aGVHD worsened in 2% (0-4%). The incidence of cGvHD within two years was 5% (1-9%). Dara had been stopped due to adverse events in 10% (95% CI 5-16%) by 24 months. At the same timepoint, 70% (60-79%) of patients had stopped because of progression. The best response to Dara was sCR/CR in 11%, VGPR in 12%, PR in 25%, SD/MR in 20% and progression in 33%. The best response was obtained at a median of 81 days (min-max 7-851 days) after starting Dara. The proportion of at least stable disease was higher when DLI treatment (n=37) was given pre-Dara. The median follow-up from the first dose of Dara was 26.8 months (95% CI 22.3 to 31.1). After starting Dara, the median PFS was 6.5 months, the median TTNT 19.3 months and median OS 21.6 months. Extra-medullary progression post-Dara was observed in 43% of patients for whom there was available data. Bone plasmacytomas were reported in 63%, soft tissues in 33% and both in 4% of cases. In total 13% of patients received a median of two DLI after starting Dara. 47% of patients received other anti-myeloma medications after Dara and 26% received radiotherapy. CONCLUSIONS The use of Dara post-allo-HSCT resulted in stable disease or better in 67% of patients. As reported previously, infections appeared to be common. Compared to the recently published data from Nikolaenko et al, there were fewer cases of aGVHD post-Dara in this retrospective analysis. The PFS were similar in both studies (4.5 vs. 6.5 months) as well as OS (17,4 v 21,6 months). A high proportion of 43% extra-medullary disease progression was observed in the current study which was not reported in the only similar study. Based on these data, Dara treatment for relapsing patients after allo-HCT creates no safety concerns and provides acceptable efficacy Disclosures Vincent: Celgene: Membership on an entity's Board of Directors or advisory committees, Other: Congress support; takeda: Membership on an entity's Board of Directors or advisory committees, Other: Congress support; janssen: Membership on an entity's Board of Directors or advisory committees, Other: Congress support. Minnema:Kite, a Gilead Company: Speakers Bureau; Celgene: Other: travel support, Research Funding; Amgen: Consultancy; Servier: Consultancy. Teipel:janssen: Honoraria. Haenel:Amgen, Novartis, Roche, Celgene, Takeda, Bayer: Honoraria. Forcade:JAZZ: Other: Travel grant for congress; NEOVII: Other: Travel grant for congress; Gilead: Speakers Bureau; Sanofi: Other: Travel grant for congress; Novartis: Other: Travel grant for congress. Schönland:Janssen, Prothena, Takeda: Honoraria, Other: travel support to meetings, Research Funding. Yakoub-Agha:Celgene: Honoraria; Novartis: Honoraria; Gilead/Kite: Honoraria, Other: travel support; Janssen: Honoraria; Jazz Pharmaceuticals: Honoraria. Beksac:Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Sanofi: Speakers Bureau; Amgen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen&janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Deva: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
- Published
- 2020
8. Triangle: Autologous Transplantation after a Rituximab/Ibrutinib/ara-c Containing Induction in Generalized Mantle Cell Lymphoma - a Randomized European MCL Network Trial
- Author
-
Gregor Verhoef, Arne Kolstad, Martin Dreyling, Michal Szymczyk, Wolfram Klapper, Ulrich Mey, Jeanette K. Doorduijn, Christiane Pott, Eva Hoster, Eva Giné, Martin Hutchings, Mats Jerkeman, Maria Gomes da Silva, Marek Trneny, Michael Unterhalt, and Marco Ladetto
- Subjects
Melphalan ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Lymphoma ,chemistry.chemical_compound ,chemistry ,Cancer immunotherapy ,Ibrutinib ,medicine ,Cancer research ,Cytarabine ,Autologous transplantation ,Rituximab ,Mantle cell lymphoma ,business ,medicine.drug - Abstract
Background: Mantle cell lymphoma (MCL) is a distinct subtype of lymphoma with a wide variation of clinical course. Based on randomized trials of our network, current standard of care is a cytarabine-containing immunochemotherapy induction (Hermine, Lancet 2016) followed by autologous stem cell transplantation (SCT; Zöllner, ICML 2019) and rituximab maintenance for 3 years (Le Gouill, NEJM 2018). In relapsed MCL the BTK inhibitor ibrutinib achieves high response rates and ongoing remissions (Wang, NEJM 2013; Dreyling, Lancet 2016). This approach achieved especially longer remission durations in earlier treatment lines (Rule, Hamatologica 2019). We aim to clarify whether ibrutinib added to induction and as maintenance with or without autologous stem cell transplantation might improve outcome. Study design and methods: In this international, randomized three-arm phase III trial (EudraCT-no. 2014-001363-12) young, fit patients ( up to 65 years) with histologically confirmed, untreated mantle cell lymphoma advanced stage II-IV qualify for 1:1:1 randomization after written informed consent according to ICH/EU GCP. In the control arm A, patients receive an alternating R-CHOP/R-DHAP induction followed by myeloablative consolidation (either BEAM or THAM: TBI, high dose Ara-C and melphalan). In arm A+I Ibrutinib is added to the R-CHOP cycles (560 mg day 1-19) and applied as maintenance (continuous dosing) for 2 years. In arm I the same induction and maintenance is applied but high dose consolidation and autologous SCT is skipped. A rituximab maintenance (single doses every 2 months up to 3 years) may be added in all study arms according to national clinical routine. The primary study aim is to show superiority of one of three study arms as future standard of care based on the comparison of the investigator-assessed failure-free survival (FFS), i.e. to investigate if the addition of ibrutinib improves the efficacy of standard 1st line treatment, and can even challenge the use of high-dose chemotherapy with autologous SCT. Secondary study aims include the efficacy of the three treatment arms and the safety and tolerability of ibrutinib during induction immuno-chemotherapy and maintenance. Accordingly, overall and complete response rates, progression-free and overall survival will be determined as well as adverse events during induction immuno-chemotherapy and follow-up including the cumulative incidence rates of SPMs. In addition, minimal residual disease is regularly determined based on patient-specific PCR assay according to the standardized Biomed-2 procedure. Results: As of July 30th, 511 of up to 870 patients have been randomized from 12 different European countries. In a meanwhile completed safety run-in of the initial 50 patients, feasibility of the two experimental arms was confirmed with no major differences in hematological and other toxicities and no major delays during induction. Disclosures Dreyling: Acerta: Other: Scientific advisory board; Novartis: Other: Scientific advisory board; Mundipharma: Other: Scientific advisory board, Research Funding; Janssen: Other: Scientific advisory board, Research Funding, Speakers Bureau; Gilead: Other: Scientific advisory board, Speakers Bureau; Celgene: Other: Scientific advisory board, Research Funding, Speakers Bureau; Bayer: Other: Scientific advisory board, Speakers Bureau; Sandoz: Other: Scientific advisory board; Roche: Other: Scientific advisory board, Research Funding, Speakers Bureau. Ladetto:Roche: Honoraria; AbbVie: Honoraria; J&J: Honoraria; Celgene: Honoraria; Pfizer: Honoraria, Speakers Bureau; Acerta: Honoraria, Speakers Bureau; ADC Therapeutics: Honoraria. Doorduijn:Roche: Honoraria, Research Funding. Gine:Janssen: Other: Travel expenses, Research Funding; Gilead: Other: Travel expenses, Research Funding; Roche: Other: Travel expenses, Research Funding. Jerkeman:Janssen: Honoraria, Research Funding; Gilead: Honoraria, Research Funding; Acerta: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Roche: Honoraria, Research Funding. Mey:Janssen-Cilag: Consultancy; Roche: Consultancy, Research Funding. Hutchings:Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Research Funding; Celgene: Research Funding; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Research Funding. Kolstad:Merck: Research Funding; Nordic Nanovector: Membership on an entity's Board of Directors or advisory committees, Research Funding. Trneny:Roche: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria; Incyte: Consultancy, Honoraria; Gilead sciences: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Morphosys: Consultancy, Honoraria. Gomes da Silva:AbbVie: Consultancy, Other: Travel support; Roche: Consultancy, Other: Travel support; Janssen-Cilag: Consultancy, Other: Travel support; Celgene: Consultancy; Gilead Siences: Other: Travel support, Research Funding. Klapper:Roche, Takeda, Amgen, Regeneron: Honoraria, Research Funding. Unterhalt:F. Hoffmann-La Roche: Research Funding. Hoster:Janssen: Research Funding; Roche Pharma AG: Other: Travel Support.
- Published
- 2019
9. A Multistate Survival Analysis for Patients with Follicular Lymphoma (FL) Using 13 First-Line Randomized Trials from FL Analysis of Surrogate Hypothesis (FLASH) Group
- Author
-
Michele Ghielmini, Umberto Vitolo, Wolfgang Hiddemann, Eva Kimby, Robert Marcus, Bruce A. Peterson, Jesse G. Dixon, Catherine Sebban, Emmanuel Gyan, Charles Foussard, Marco Ladetto, Michael Herold, Gilles Salles, Christopher R. Flowers, Howard S. Hochster, Tina Nielsen, Eva Hoster, Franck Morschhauser, Qian Shi, Mathias J. Rummel, Caglar Caglayan, Anna Wall, Anton Hagenbeek, and Kenneth A. Foon
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,First line ,Immunology ,Follicular lymphoma ,Cell Biology ,Hematology ,Coping behavior ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Rituximab ,Progression-free survival ,business ,Survival analysis ,medicine.drug - Abstract
Introduction: Most patients with newly diagnosed FL treated with rituximab (R) alone or R + chemotherapy will experience prolonged progression-free survival and overall survival (OS), but it remains unclear what factors have the greatest influence on FL-associated and other causes of death in this patient population. We utilized individual patient data from 13 first-line randomized clinical trials from the FLASH database to perform a comprehensive multistate survival analysis to examine and quantify the relationships between clinical characteristics, treatment response, and early, intermediate, and late FL outcomes. Methods: The multistate survival analysis model defined states for "Alive after beginning Induction Treatment (TX)", "Alive after beginning Maintenance TX", "Death due to FL", and "Death from Other Causes" (Figure 1). We used the Aalen-Johansen estimator, a generalization of the Kaplan-Meier estimator, to calculate the likelihood of being in each model state and estimate the course of FL over time. Making no assumptions on the probability distributions and capable of coping with censored observations, the Aalen-Johansen estimator is a convenient and reliable nonparametric estimator for clinical data. Results: Among 7,465 FL patients with median age 56 (range 18-90) years, 49.2% were female; 28.7% Stage I-III, 71.3% Stage IV, and FLIPI was 0-1 (20.0%), 2 (36.8%), ≥ 3 (43.2%). Following initiation of induction treatment, 2-, 5- and 10-year death rates were 1.7%, 3.8%, and 5.8% due to FL, and 0.7%, 2.1%, and 4.8% from other causes (Figure 2). Death rates at 2, 5, and 10 years due to FL and other causes for subgroups based on clinical characteristics and treatment response are shown in Table 1. Notably, patients > 70 years and patients with FLIPI ≥ 3 had worse outcomes and patients achieving CR at 18, 24, and 30 months experienced improved outcomes. Conclusion: This is the largest study using data from randomized trials to quantify the impact of clinical factors on early, intermediate and late mortality by cause of death. We demonstrated that age > 70 years and FLIPI ≥ 3 were linked to increased FL-associated death and response to TX distinguished patients with favorable and poor outcomes. Future analyses should quantify the impact of predictors on the rate/time of FL outcomes in multivariable models. Disclosures Salles: Roche, Janssen, Gilead, Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Educational events; Amgen: Honoraria, Other: Educational events; BMS: Honoraria; Merck: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis, Servier, AbbVie, Karyopharm, Kite, MorphoSys: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Educational events; Epizyme: Consultancy, Honoraria; Autolus: Consultancy, Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Educational events. Hoster:Janssen: Research Funding; Roche Pharma AG: Other: Travel Support. Hiddemann:Bayer: Research Funding; Gilead: Consultancy, Honoraria; Vector Therapeutics: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Research Funding; Roche: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria. Herold:Roche: Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Gilead: Honoraria; Celgene: Honoraria. Morschhauser:Servier: Consultancy; Gilead: Consultancy; Janssen: Honoraria; Roche/Genentech: Consultancy; BMS: Honoraria; Celgene: Honoraria. Rummel:Roche Pharma AG: Honoraria, Research Funding; Celgene: Honoraria; Janssen: Honoraria; Sandoz: Honoraria. Kimby:AbbVie,: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Roche: Membership on an entity's Board of Directors or advisory committees; Jansen: Membership on an entity's Board of Directors or advisory committees; Gilead: Other: educational lectures. Vitolo:Abbvie: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Kite: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Gilead: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Juno Therapeutics: Membership on an entity's Board of Directors or advisory committees; F. Hoffmann-La Roche: Speakers Bureau; Novartis: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Gyan:Pfizer: Honoraria. Ladetto:Celgene: Honoraria; Roche: Honoraria; Janssen: Honoraria; Abbvie: Honoraria; Acerta: Honoraria; Sandoz: Honoraria. Nielsen:F. Hoffmann-La Roche Ltd: Employment, Equity Ownership. Flowers:AstraZeneca: Consultancy; BeiGene: Consultancy, Research Funding; Eastern Cooperative Oncology Group: Research Funding; Burroughs Wellcome Fund: Research Funding; AbbVie: Consultancy, Research Funding; Optimum Rx: Consultancy; V Foundation: Research Funding; Pharmacyclics/Janssen: Consultancy, Research Funding; Bayer: Consultancy; Gilead: Consultancy, Research Funding; TG Therapeutics: Research Funding; Denovo Biopharma: Consultancy; Acerta: Research Funding; National Cancer Institute: Research Funding; Millenium/Takeda: Research Funding; Genentech, Inc./F. Hoffmann-La Roche Ltd: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; Spectrum: Consultancy
- Published
- 2019
10. Definition and Validation of the New Elderly Prognostic Index (EPI) for Elderly Patients with Diffuse Large B-Cell Lymphoma Integrating Geriatric and Clinical Assessment: Results of the Prospective 'Elderly Project' on 1353 Patients By the Fondazione Italiana Linfomi
- Author
-
Benedetta Puccini, Sofia Kovalchuk, Michele Spina, Francesca Re, Federica Cavallo, Annalisa Chiappella, Chiara Bottelli, Alessandra Tucci, Maria Giuseppina Cabras, Elsa Pennese, Michele Merli, Luigi Petrucci, Gerardo Musuraca, L. Flenghi, M. Christina Cox, Luca Nassi, Vittorio Ruggero Zilioli, Anna Lia Molinari, Roberto Sartori, Valentina Tabanelli, Simone Ferrero, Stefan Hohaus, Monica Balzarotti, D Dessi, Caterina Mammi, Marco Ladetto, Francesco Angrilli, Alberto Fabbri, Stefano Luminari, Francesco Merli, Annalisa Arcari, Guido Gini, Emanuele Cencini, Monica Tani, Dario Marino, and Luigi Marcheselli
- Subjects
Geriatrics ,medicine.medical_specialty ,Index (economics) ,Palliative care ,business.industry ,Immunology ,Instrumental ADL ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,International Prognostic Index ,Family medicine ,Honorarium ,medicine ,Observational study ,business ,Diffuse large B-cell lymphoma - Abstract
Introduction: Management of elderly patients with Diffuse Large B-Cell Lymphoma (DLBCL) is challenging. A simplified Comprehensive Geriatric Assessment (sCGA) based on ADL (Activity of Daily Living), IADL (Instrumental ADL) and CIRS-G (Comorbidity Index Rating Scale for Geriatrics) scales has demonstrated to be better than clinical judgement to stratify patients' outcome but has never been included in initial assessment. To further assess the impact of sCGA on patients' outcome, we conducted a prospective observational study on a large series of elderly patients with DLBCL. Methods: Patients were enrolled if 65 year old or older, with an untreated de novo DLBCL. sCGA was available at a web based platform that classified patients as FIT, UNFIT, and FRAIL, as shown in Table 1. Treatment choice was left at physician discretion. According to anthracycline dose, therapy was classified as curative (≥70% of full anthracycline dose), intermediate ( Results: From December 2013 to December 2017, 1353 patients have been registered by 37 centres and 1207 were eligible. Median age was 76 years (65-94), 68% had stage III-IV, and 55% had an International Prognostic Index(IPI) ≥3; 500 (42%), 304 (25%), and 403 (33%) were classified as FIT, UNFIT and FRAIL, respectively. Data on treatment were available in 1164 patients: rituximab was used in 96% of patients; treatment was curative in 89%, 53%, and 36% of FIT, UNFIT, and FRAIL patients, respectively; intermediate in 10%, 39%, and 31%, palliative in 0%, 8%, and 33% of patients. The OS was available in 1158 out 1164 cases. With a median follow up of 30 months (1-59) 3y-OS was 64% (95% CI 61% to 67%). According to sCGA the OS was significantly different among the 3 geriatric groups. Correlation with OS was improved when sCGA was integrated with age < or ≥ 80 years to define 3 groups of patients (Table 2): FIT and UNFIT younger than 80 years (sCGA Group 1; 55%, 3 yr OS 75%), UNFIT ≥ 80 years and FRAIL younger than 80 years (sCGA Group 2: 28%, 3yr OS 58%), FRAIL ≥ 80 years (sCGA Group 3: 17%; 3yr OS 43%). Univariable and multivariable analysis for OS was conducted using the 3 sCGA groups and other clinical and laboratory features. The 3 sCGA groups were shown as independent prognostic factors with IPI and with anemia (Hb < 12 g/dl). We used results of multivariable analysis to build a categorical prognostic index assigning different weights to prognostic features based on their Hazard Ratio (HR) (Table 3). The Elderly Prognostic Index (EPI) was defined as the score obtained from the sum of the weights and allowed to define 3 risk groups: Low Risk (LR: score 0-1; 23% of patients); Intermediate Risk (IR; score 2-4; 48%); High Risk (HiR; score 5-7; 29%). The 3 EPI risk groups had a different 3 year OS of 87%(95%CI 81-91), 69%(95%CI 63-73), and 42% (95%CI 36-49); HR for IR vs LR 2.57 (1.72, 3.84); HiR vs LR 6.21(4.17 -9.25), HiR vs IR 2.42 (1.91-3.05) (Figure1). Regarding treatment modality, curative, intermediate and palliative therapies were adopted in 89%, 10%, and 1% of the LR group; 70%, 24%, 7% of the IR group, and 37%, 35%, 28% of the HiR group. The model was internally validated by means of 1000 procedures confirming good performance (slope shrinkage 0.935 and c-Harrell 0.675 in validation sample compared with 0.682 in training sample). The EPI was also tested in an external validation data set that was identified from the pivotal study of sCGA in DLBCL (N=172 patients, Tucci A. et al, Leuk Lymph, 2015) (Figure 1). Conclusion: Using data from this large prospective observational study on elderly DLBCL patients we were able to build a new prognostic index that allows to identify 3 risk groups with significant differences in terms of 3 years OS. The EPI is the first index that integrates geriatric assessment with clinical features and contributes to improving management and clinical research in elderly patients with DLBCL. Disclosures Spina: Servier: Membership on an entity's Board of Directors or advisory committees, Other: lecture fee; Sandoz: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Mundipharma: Membership on an entity's Board of Directors or advisory committees, Other; Roche: Other: lecture fee; Teva: Membership on an entity's Board of Directors or advisory committees, Other: lecture fee; GILEAD: Membership on an entity's Board of Directors or advisory committees, Other: lecture fee; Celgene: Other: lecture fee; BMS: Other: lecture fee; Sanofi Genzyme: Other: lecture fee; CTI: Membership on an entity's Board of Directors or advisory committees, Other: lecture fee; Menarini: Membership on an entity's Board of Directors or advisory committees, Other: lecture fee, Research Funding; Takeda: Other: lecture fee; Janssen-Cilag: Membership on an entity's Board of Directors or advisory committees, Other: lecture fee; Pfizer: Membership on an entity's Board of Directors or advisory committees. Merli:Janssen: Honoraria; Takeda: Honoraria, Other: Travel Expenses; Gilead: Honoraria; Mundipharma: Honoraria; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel expenses, Research Funding; Sandoz: Membership on an entity's Board of Directors or advisory committees; Teva: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees, Other: Travel Expenses. Cavallo:Takeda: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Ladetto:Roche: Honoraria; AbbVie: Honoraria; J&J: Honoraria; Celgene: Honoraria; ADC Therapeutics: Honoraria; Acerta: Honoraria, Speakers Bureau; Pfizer: Honoraria, Speakers Bureau. Chiappella:Celgene: Other: advisory board, Speakers Bureau; Janssen: Other: advisory board, Speakers Bureau; Servier: Other: advisory board, Speakers Bureau; Roche: Speakers Bureau; Teva: Speakers Bureau. Nassi:Takeda: Consultancy; Janssen: Consultancy; Merck: Consultancy. Ferrero:Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Gilead: Speakers Bureau; Servier: Speakers Bureau; EUSA Pharma: Membership on an entity's Board of Directors or advisory committees. Luminari:ROCHE: Membership on an entity's Board of Directors or advisory committees; CELGENE: Membership on an entity's Board of Directors or advisory committees, Other: Travel Grant; GILEAD: Other: Lecturer; TAKEDA: Other: Travel Grant.
- Published
- 2019
11. EARLY STAGE Follicular Lymphoma: First Results of the FIL 'Miro' Study, a Multicenter Phase II Trial Combining Local Radiotherapy and MRD-Driven Immunotherapy
- Author
-
Alessandro Pulsoni, Valter Gattei, Sara Galimberti, Antonella Anastasia, Monica Tani, Tommasina Perrone, Patrizia Bernuzzi, Giovanni Partesotti, Marzia Cavalli, Carola Boccomini, Lucia Anna De Novi, Clara Mannarella, Luca Nassi, Caterina Stelitano, Marco Ladetto, Natalia Cenfra, Giorgia Annechini, Sara Rattotti, Emanuele Cencini, Maria Elena Tosti, Anna Lia Molinari, Barbara Mantoan, Luca Arcaini, Paolo Corradini, Daniela Renzi, Elena Ciabatti, Anna Guarini, Robin Foà, Silvia Bolis, Irene Della Starza, Ilaria Del Giudice, Vittorio Ruggero Zilioli, Stefano Luminari, Andrés J.M. Ferreri, Gerardo Musuraca, Giovanni Manfredi Assanto, Francesca Re, Simone Ferrero, Anna Marina Liberati, Umberto Ricardi, Donato Mannina, Lavinia Grapulin, and Emanuela Zanni
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Chronic lymphocytic leukemia ,Immunology ,Follicular lymphoma ,Phases of clinical research ,Cell Biology ,Hematology ,Immunotherapy ,medicine.disease ,Ofatumumab ,Biochemistry ,Radiation therapy ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,Combined Modality Therapy ,Stage (cooking) ,business - Abstract
Introduction Limited stage follicular lymphoma (FL) is usually managed with involved field radiotherapy (IFRT), although different approaches are currently carried out, ranging from watch and wait to combined treatment. RT on involved lymph nodes allows eradication of the disease only in 40-50% of patients. Anti-CD20 monoclonal antibodies (MoAb), widely used in advanced stage FL, are likely to be effective in reducing the relapse risk, although no scientific evidence of their role has been provided. The aim of this multicenter phase II prospective study was to evaluate the role of MRD in identifying patients unlikely to be cured by RT, for whom an immunotherapy-based consolidation could improve outcome. Methods 110 patients with stage I/II FL were enrolled. IFRT was administered to all patients at a dose of 24 Gy. Peripheral blood (PB) and bone marrow (BM) samples were centralized to the Italian FIL (Federazione Italiani Linfomi) MRD Network of EuroMRD-certified laboratories: the presence of a BCL2/IGH rearrangement was investigated at baseline in all patients by nested PCR (NEST) and RQ-PCR (RQ), the latter according to the EuroMRD guidelines. In patients BCL2/IGH+ at baseline by both NEST and RQ in BM and/or PB, MRD was analyzed in both tissues after IFRT and every 6 months over a three-year follow-up period. Patients with positive MRD by both NEST and RQ in BM and/or PB after IFRT or who became positive during the follow-up were treated with 8 weekly doses of the anti-CD20 MoAb ofatumumab. The primary objective of the study was to define the efficacy of immunotherapy in obtaining the disappearance of BCL2/IGH rearranged cells. Results Preliminary data are available for 107 patients, 57 males, 50 females. Median age was 55 years (29-83). 17% had G1 FL, 32% G2, 40% G3A, 11% NOS. The FLIPI score was 0 in 59% of patients, 1 in 35%, 2 in 6%. 69% of patients had inguinal site involvement. Despite a negative BM biopsy, at baseline 30% of patients (n=32) had a BCL2/IGH rearrangement (30 MBR, 1 MBR and mcr, 1 mcr) in the BM and/or PB; the concordance between compartments was 90%, with 10% of negative PB showing a positive BM. No significant differences were observed in relapse probability between patients with or without a molecular marker. All patients were submitted to IFRT and all obtained a clinical response, which was complete in 79 of the 101 evaluated patients (78%) and partial in 22 (22%). MRD evaluation after treatment revealed the persistence of BCL2/IGH rearranged cells in the PB and/or BM in 60% of patients. According to the design of the protocol, MRD-positive patients, either after IFRT (n=18) or in case of conversion to a positive signal during the follow-up (n=7), received 8 weekly administration of ofatumumab. A conversion to MRD negativity, evaluated in 23 treated patients, was obtained in 20 (87% - CI 65.1-97.1). This result was significantly superior to the expected 50%. One death occurred after IFRT, due to ischemic stroke. Adverse events likely correlated to ofatumumab occurred in 7/25 treated patients, consisting of infusion reactions in 5, leading to a permanent interruption of immunotherapy in 3. After a median follow-up of 18 months, all patients who achieved a MRD negativity with ofatumumab underwent a regular molecular follow-up and are still MRD-negative. Overall, clinical relapse or progression were observed in 17 patients: 13 (18%) among the 73 "no marker" patients; 2 relapses (16%) were observed among the 12 MRD-negative patients after IFRT and 2 relapses were observed among the 23 patients treated with the anti-CD20 MoAb (8.7%), 1 having achieved a MRD negativity and 1 not. No significant differences in event-free survival have so far been observed between the three groups. Conclusions The MRD data of this phase II trial for early stage FL indicate that RT alone is often insufficient to eradicate the disease, being capable of inducing a negative MRD only in 40% of evaluable cases, with a long-lasting effect only in half of them. The primary objective of this study - MRD negativity after immunotherapy - was achieved, obtaining the disappearance of BCL2/IGH rearranged cells in the majority of patients treated with ofatumumab. The strategy of an immunotherapy consolidation after IFRT in MRD-positive patients allowed to increase molecular responses. A longer follow-up and further studies on larger patient populations will allow to conclusively define the impact of this MRD-driven strategy also on clinical outcome. Disclosures Pulsoni: Roche: Consultancy, Speakers Bureau; Takeda: Consultancy; Pfizer: Consultancy; Sandoz: Consultancy; Gilead: Speakers Bureau; Merk: Consultancy; Bristol Meyer Squibb: Speakers Bureau. Ferrero:Servier: Speakers Bureau; EUSA Pharma: Membership on an entity's Board of Directors or advisory committees; Gilead: Speakers Bureau; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Luminari:ROCHE: Other: Role as Advisor ; CELGENE: Other: Role as Advisor & Travel Grant; TAKEDA: Other: Travel Grant; GILEAD: Other: Lecturer . Liberati:Amgen: Membership on an entity's Board of Directors or advisory committees, Other: Clinical trial support; Celgene: Honoraria, Other: Clinical trial support; Bristol-Myers Squibb: Honoraria; Takeda: Membership on an entity's Board of Directors or advisory committees; Incyte: Consultancy; Janssen: Honoraria; Servier: Honoraria, Membership on an entity's Board of Directors or advisory committees; AbbVie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Clinical trial support; Roche: Other: Clinical trial support; Novartis: Other: Clinical trial support. Ferreri:Roche: Research Funding; Celgene: Consultancy, Research Funding; Novartis: Consultancy; Kite: Consultancy. Nassi:Takeda: Consultancy; Janssen: Consultancy; Merck: Consultancy. Corradini:Roche: Honoraria; Novartis: Honoraria; kite: Honoraria; KiowaKirin: Honoraria; Janssen: Honoraria; Gilead: Honoraria; Daiichi Sankyo: Honoraria; Celgene: Honoraria; Amgen: Honoraria; Abbvie: Honoraria; Servier: Honoraria; Sanofi: Honoraria; Takeda: Honoraria. Mannina:Janssen: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees. Arcaini:Celgene: Speakers Bureau; Gilead Sciences: Research Funding; Bayer, Celgene, Gilead Sciences, Roche, Sandoz, Janssen-Cilag, VERASTEM: Consultancy; Celgene, Roche, Janssen-Cilag, Gilead: Other: Travel expenses. Galimberti:Roche: Speakers Bureau; Celgene: Speakers Bureau; Novartis: Speakers Bureau. Ladetto:AbbVie: Honoraria; Roche: Honoraria; ADC Therapeutics: Honoraria; Acerta: Honoraria, Speakers Bureau; Pfizer: Honoraria, Speakers Bureau; J&J: Honoraria; Celgene: Honoraria. Foà:Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Abbvie: Consultancy, Speakers Bureau; Roche: Consultancy, Speakers Bureau; Roche: Consultancy, Speakers Bureau; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celltrion: Membership on an entity's Board of Directors or advisory committees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Shire: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Amgen Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Abbvie: Consultancy, Speakers Bureau; Celltrion: Membership on an entity's Board of Directors or advisory committees; Amgen Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Shire: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. OffLabel Disclosure: The anti-CD20 MoAb Ofatumomab is employed to eradicate Minimal Residual Disease in early stage Follicular Lymphoma(FL). The drug is registered for Chronic Lymphocytic Leukemia, not for FL.
- Published
- 2019
12. Outcomes Following Second Allogenic Haematopoietic Cell Transplantation in Patients with Myelofibrosis: A Retrospective Study on Behalf of the Chronic Malignancies Working Party of EBMT
- Author
-
Martin Bornhäuser, Liesbeth C. de Wreede, Fabio Ciceri, Mitja Nabergoj, Nicolaus Kröger, Uwe Platzbecker, Donal P. McLornan, Henrik Sengeloev, Tomasz Czerw, Linda Koster, Yves Chalandon, Peter Dreger, Emanuele Angelucci, Marie Robin, Stephen D. Robinson, Marco Ladetto, Matthias Stelljes, Ibrahim Yakoub-Agha, Alessandro Rambaldi, Xavier Poiré, Juan Carlos Hernandez Boluda, Jakob Passweg, Jiri Mayer, Junfeng Wang, and Patrick Hayden
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Graft failure ,Graft rejection ,Karnofsky Performance Status ,business.industry ,Immunology ,Haematopoietic cell transplantation ,Retrospective cohort study ,Cell Biology ,Hematology ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Curative treatment ,Family medicine ,Honorarium ,Medicine ,In patient ,business ,health care economics and organizations ,030215 immunology - Abstract
Introduction: The only curative treatment for myelofibrosis (MF) remains allogenic haematopoietic cell transplantation (allo-HCT) although the risks of non-relapse mortality (NRM), relapse and graft rejection need to be taken into consideration. Therapeutic approaches following relapse after allo-HCT include symptom-directed management, chemotherapy, JAK2 inhibitors, adoptive immunotherapeutic approaches with donor lymphocyte infusion (DLI) and, in a minority, a second allo-HCT. Frequently, due to the advanced age of the recipient, early relapses, and numerous complications, 2nd allo-HCT can only be considered in a limited number of patients. Few studies evaluating the role of 2nd allo-HCT in MF following 1st relapse or primary/secondary graft rejection have been published to date. Methods and results: Patient selection was performed by identifying adult patients who underwent 2nd allo-HCT for MF between 2010-2017: 216 patients were analyzed; 64% were male, 78% had primary MF (PMF) and 22% secondary MF (sMF). Median age at the time of 2nd allo-HCT was 57 years, and median time from 1st allo-HCT was 8 months. Of this cohort, 56% of patients received a 2nd allo-HCT for relapse, 31% for graft failure and the reason was missing in 13%. A greater proportion was transplanted within 1 year from 1st allo-HCT (61 %) whilst 39% had 2nd allo-HCT > 1 year. A reduced Karnofsky performance status (KPS12 months, p=0.02). The 2-year relapse-free-survival (RFS) for the entire cohort was 44%. Only time elapsed from the 1st allo-HCT to 2nd was significantly associated with 2-year RFS (41% for ≤12 months, 49% for >12 months, p=0.05). Of note, the 2-year OS and RFS were comparable following use of the same or a different donor. The 2-year cumulative incidence of relapse and NRM were 22 and 34%, respectively. The time interval from 1st to 2nd allo-HCT appeared to be highly significant for NRM with patients transplanted ≤12 months having a higher 2-year NRM compared to those transplanted >12 months (40 vs 24%, respectively, p=0.008). A trend for higher NRM was the reason for 2nd allo-HCT: patients transplanted for graft rejection had a 2-year NRM of 45% compared to 31% for those with relapse (p=0.06). Conclusions: This analysis supports the utilization of a 2nd allo-HCT for patients with MF who have presented with graft failure or relapse following a 1st allo-HCT. In univariate analysis, overall outcome appears worse in patients being transplanted after graft failure as well as for those transplanted within 1 year after 1st allo-HCT, due to increased NRM. Of note, the use of either the original or a different donor are associated with similar outcomes. Further work is required to elucidate other risk factors, GVHD rates and to define the optimal conditioning regimen in this setting. Table. Disclosures Robin: Novartis Neovii: Research Funding. Kröger:Sanofi-Aventis: Honoraria; Riemser: Research Funding; Novartis: Honoraria, Research Funding; Neovii: Honoraria, Research Funding; Medac: Honoraria; JAZZ: Honoraria; DKMS: Research Funding; Celgene: Honoraria, Research Funding. Angelucci:Celgene: Honoraria, Other: Participation in DMC; BlueBirdBio: Other: Local advisory board; Jazz Pharmaceuticals: Other: Local advisory board; Roche: Other: Local advisory board; Vertex Pharmaceuticals Incorp., and CRISPR Therapeutics: Other: Participation in DMC; Novatis: Honoraria, Other: Chair Steering Committee TELESTO protocol. Dreger:AbbVie, AstraZeneca, Gilead, Janssen, Novartis, Riemser, Roche: Consultancy; AbbVie, Gilead, Novartis, Riemser, Roche: Speakers Bureau; MSD: Membership on an entity's Board of Directors or advisory committees, Other: Sponsoring of Symposia; Neovii, Riemser: Research Funding. Platzbecker:Celgene: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Abbvie: Consultancy, Honoraria. Rambaldi:Novartis: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Speakers Bureau; Amgen: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Research Funding, Speakers Bureau; Italfarmaco: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Research Funding, Speakers Bureau; Gilead: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Speakers Bureau; Roche: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Research Funding, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Other: travel support, Speakers Bureau; Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Jazz: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Omeros: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Mayer:AOP Orphan Pharmaceuticals AG: Research Funding. Ladetto:ADC Therapeutics: Honoraria; Pfizer: Honoraria, Speakers Bureau; Celgene: Honoraria; J&J: Honoraria; Roche: Honoraria; AbbVie: Honoraria; Acerta: Honoraria, Speakers Bureau. Hernandez Boluda:Incyte: Other: Travel expenses paid. McLornan:Jazz Pharmaceuticals: Honoraria, Speakers Bureau; Novartis: Honoraria. Chalandon:Incyte Biosciences: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria.
- Published
- 2019
13. Comprehensive Minimal Residual Disease (MRD) Analysis of the Fondazione Italiana Linfomi (FIL) MCL0208 Clinical Trial for Younger Patients with Mantle Cell Lymphoma: A Kinetic Model Ensures a More Refined Risk Stratification
- Author
-
Elisa Genuardi, Claudia Castellino, Giovannino Ciccone, Manuela Zanni, Gerardo Musuraca, Luigia Monitillo, Vincenzo Pavone, Umberto Vitolo, Pietro Maria Stefani, Nicola Cascavilla, Marco Ladetto, Sergio Cortelazzo, Simone Ferrero, Stefan Hohaus, Daniele Grimaldi, Annalisa Cifaratti, Francesca Re, Anna Marina Liberati, Ivana Casaroli, Mario Petrini, Mariella Lo Schirico, Gian Maria Zaccaria, Fabio Benedetti, Andrea Evangelista, Daniela Drandi, Angela Congiu, and Barbero Daniela
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Kinetic model ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Minimal residual disease ,Peripheral blood ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Outcome predictor ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,Risk stratification ,medicine ,Mantle cell lymphoma ,business ,Lenalidomide ,medicine.drug - Abstract
Background and Aims. Minimal residual disease (MRD) detection by PCR-based methods is a relevant outcome predictor in MCL, however it is not clear which might represent the most effective methodology (nested vs real-time quantitative PCR, RQ-PCR), the most informative tissue source (bone marrow, BM, vs peripheral blood, PB), the best timing of analysis (midterm vs post-therapy) and the added value of performing multiple MRD determinations. To address these issues a systematic MRD detection program was performed in the Fondazione Italiana Linfomi (FIL) MCL0208 trial (NCT02354313), a prospective, randomized phase III trial comparing lenalidomide maintenance vs observation after an intensive citarabine containing chemo-immunotherapy (R-HDS) program followed by ASCT in 300 frontline MCL patients Disclosures Vitolo: Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Sandoz: Speakers Bureau; Gilead: Speakers Bureau; Takeda: Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.
- Published
- 2018
14. Lenalidomide Maintenance after Autologous Transplantation Prolongs PFS in Young MCL Patients: Results of the Randomized Phase III MCL 0208 Trial from Fondazione Italiana Linfomi (FIL)
- Author
-
Alessandro Re, Michael Mian, Sergio Cortelazzo, Alberto Zamò, Maria Gomes da Silva, Umberto Vitolo, Armando Santoro, Vittorio Stefoni, G. Ciccone, Filippo Ballerini, Simone Ferrero, Andrea Evangelista, Manuel Gotti, Marco Ladetto, Angela Coggi, Annalisa Chiappella, Chiara Rusconi, Franco Narni, Andrés J.M. Ferreri, Alberto Bosi, Maurizio Martelli, Alice Di Rocco, Giuseppe Rossi, Anna Lia Molinari, Caterina Stelitano, and Federica Cavallo
- Subjects
education.field_of_study ,medicine.medical_specialty ,Study drug ,business.industry ,Immunology ,Population ,Cell Biology ,Hematology ,Biochemistry ,03 medical and health sciences ,Regimen ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Family medicine ,medicine ,Clinical endpoint ,Autologous transplantation ,Elevated ldh ,Stage iv ,education ,business ,030215 immunology ,Lenalidomide ,medicine.drug - Abstract
Background. Ara-c based chemo-immunotherapy followed by autologous stem cell transplantation (ASCT) is the most effective approach in young mantle cell lymphoma (MCL) patients, though few if any patients are cured. Recent data indicate that subsequent Rituximab maintenance (RM) prolongs PFS and OS (Le Gouill NEJM 2017). Lenalidomide is an oral agent effective in MCL, considered suitable for prolonged maintenance programs, but has never been tested in this setting. The FIL MCL0208 trial (NCT02354313) is a prospective, international randomized, phase III trial, comparing Lenalidomide maintenance (LM) vs observation (OBS) after an intensive Ara-c containing chemo-immunotherapy (R-HDS) program, followed by ASCT in previously untreated MCL patients. Patients and Methods. Adult patients aged 18-65 years, with advanced stage MCL without clinically significant comorbidities were enrolled. Patients received 3 R-CHOP-21, followed by R-HDS i.e. R-high-dose Cyclophosphamide (R-HD-CTX) (4g/m2), 2 cycles of R-high-dose Ara-C (R-HDAC) (2g/m2 q12x3 d). CD34+ cells were collected after the first course of R-HDAC. The conditioning regimen for ASCT was BEAM. After ASCT, responding patients were randomized between LM (15 mg days 1-21 every 28 days) for 24 months or observation. Primary endpoint analysis was scheduled at the occurrence of the 60th PFS event in the randomized population, which occurred on June 20th, 2017 and data were analyzed for the present abstract on March 3rd 2018. Results. Three-hundred three patients were enrolled from May 2008 to August 2015 by 48 Italian and 1 Portuguese Center. Three patients were excluded after central histological review. Median age was 57 years (IQR 51-62), M/F ratio 3.6/1. Ninety-two percent of patients had stage IV, 33% bulky disease (>5 cm), 33% elevated LDH, and 75% BM infiltration. Ki67 ≥30% was observed in 32%, MIPI was low (L) in 54%, intermediate in 31% and high (H) in 15% of patients. MIPI-c was L in 49%, low-intermediate (LI) in 29%, high-intermediate (HI) in 14%, H in 9%. Nine percent had blastoid variant. Fifty-two (17%) patients interrupted treatment before randomization (8 toxic deaths, 1 death for car accident, 24 progressions and 19 toxicity/refusals). On an ITT basis, the R-HDS + ASCT program induced 78% of CR, 7% of PRs, 10% of PD, 3% of toxic deaths (TRM) and 2% NA. Median follow-up (mFU) from inclusion was 51 months. Three years PFS and OS for the enrolled population were 67% and 84%, respectively. Of 248 patients who received ASCT, 205 were randomized either to LM (n=104) or OBS (n=101) and 43 (17%) were not because of: lack of response (8), refusal/PI decision/delay (8), unresolved infections (3) and inadequate hematopoietic recovery (24). Feasibility and efficacy were assessed on an ITT basis while toxicity was analyzed on subjects receiving at least one Lenalidomide dose. In the LM arm, 53 out of 104 patients did not start or complete the planned maintenance because of death (2), AE (26), PD (7), still ongoing (2), other causes (16). In the OBS arm 32 patients did not complete the observation phase because of death (1), AE (1), PD (20), still ongoing (10), other causes (1). Overall 28% of patients received less than 25% of the planned Lenalidomide dose. Despite suboptimal exposure to study drug, with a mFU from randomization of 35 months, 22 PFS events were recorded in the LM cohort vs 38 in the OBS arm, resulting in a 3y-PFS of 80% (95% CI; 70%-87%) in the LM arm vs. 64% in the OBS arm (95% CI; 53%-73%), stratified HR 0.51; 95% CI 0.30-0.87; p=0.013 (Fig 1A). OS was superimposable in the two arms: 93% vs 86%, stratified HR 0.96, 95% CI 0.44-2.11, p= 0.91 (Fig1B). Two deaths were observed in the LM arm due to pneumonia and thrombotic thrombocytopenic purpura and one in the OBS arm due to pneumonia. Grade 3-4 hematological toxicity was seen in 63% of patients in LM vs 11% in the OBS arm with 59% vs 10% of patients experiencing granulocytopenia. Non-hematological grade 3 toxicity was comparable in the two arms except grade 3-4 infections (11% vs. 4%; Fisher's p=0.10). Second cancers occurred in 7 patients in the LM and 3 in the OBS arm (Fisher's p=0.20). Conclusions. Results from the MCL0208 trial indicate that LM has a clinically meaningful anti-lymphoma activity in MCL. However, the applicability of LM has some limitations in the context of patients undergoing intensified chemoimmunotherapy. Overall these data support the use of a maintenance regimen after ASCT in young MCL patients. Disclosures Ladetto: Roche: Honoraria; Celgene: Honoraria; Acerta: Honoraria; Jannsen: Honoraria; Abbvie: Honoraria; Sandoz: Honoraria. Di Rocco:Roche: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Rossi:Novartis: Honoraria; Jazz: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees; Teva: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees, Other: Travel expenses; Amgen: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees, Other: Travel expenses; Janssen: Membership on an entity's Board of Directors or advisory committees, Travel expenses; Roche: Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria; Mundipharma: Honoraria; Sandoz: Honoraria; Seattle Genetics: Research Funding; Alexion: Other: Travel expenses. Chiappella:Janssen: Membership on an entity's Board of Directors or advisory committees, Other: lecture fees; Roche: Other: lecture fees; Teva: Other: lecture fees; Nanostring: Other: lecture fees; Amgen: Other: lecture fees; Celgene: Membership on an entity's Board of Directors or advisory committees, Other: lecture fees. Rusconi:Celgene: Research Funding. Gomes da Silva:Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees, Institution's payment for consultancy, Travelling support; Celgene: Other: Travelling support; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees; Roche: Other: Institution's payment for consultancy, Travelling support; Gilead Sciences: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees, Research Funding. Vitolo:Takeda: Speakers Bureau; Sandoz: Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Gilead: Speakers Bureau. Martelli:Sandoz: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; F. Hoffman-La Roche: Membership on an entity's Board of Directors or advisory committees; Servier: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Mundipharma: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2018
15. Molecular Subtypes of Splenic Marginal Zone Lymphoma (SMZL) Are Associated with Distinct Pathogenic Mechanisms and Outcomes - Interim Analysis of the IELSG46 Study
- Author
-
Alessandro Broccoli, Sascha Dietrich, Stefano Pileri, Maria Joao Baptista, Fabio Facchetti, Paolo Corradini, Maurilio Ponzoni, Umberto Vitolo, Manuela Mollejo, Véronique Meignin, Elena Sabattini, Alexandar Tzankov, Marco Frigeni, Juan F. García, Sílvia Beà, Francesco Passamonti, Pier Luigi Zinzani, Marco Ladetto, Carlos Montalbán, Franco Cavalli, Alessandra Tucci, Maria Gomes da Silva, Corrado Tarella, Miguel A. Piris, Adalgisa Condoluci, Gilles Salles, Carlo Visco, Govind Bhagat, Laurence de Leval, Valeria Spina, Sergio Cogliatti, Marco Paulli, Gustavo Tapia, Elias Campo, Francesca Guidetti, Luciano Cascione, Giorgio A. Vanini, Stefano Pizzolitto, Liliana Devizzi, Gianluca Gaidano, Urban Novak, Davide Rossi, Elisa Santambrogio, Estella Matutes, Emanuele Zucca, Giorgio Inghirami, Renzo Boldorini, Felicitas Hitz, Vincenzo Canzonieri, Julia T. Geyer, Gabriela Forestieri, Roberto Marasca, Antonino Maiorana, Michele Merli, Catherine Thieblemont, Alexandra Traverse-Glehen, David Oscier, Alessio Bruscaggin, Francesco Piazza, Lodovico Terzi di Bergamo, Luca Arcaini, Alessandro Rambaldi, Francesco Bertoni, and Francesco Zaja
- Subjects
0301 basic medicine ,education.field_of_study ,medicine.medical_specialty ,Splenic Marginal Zone B-Cell Lymphoma ,Immunology ,Population ,Cell Biology ,Hematology ,Gene mutation ,Biochemistry ,Actuarial survival ,Large sample ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Family medicine ,Political science ,medicine ,education ,Medical therapy ,Protein p53 - Abstract
Introduction. The majority of SMZLs display an indolent course, however the disease is still incurable and a significant proportion of patients (~25-30%) experience poor outcomes surviving 5 years, and for whom tumor material collected before initiation of medical therapy was available. Mutation analysis was performed by CAPP-seq targeted deep next generation sequencing of tumor genomic DNA. A stringent bioinformatic pipeline was applied to suppress the background noise allowing to call variants with a sensitivity of 5x10-2 in FFPE derived DNA. Copy number variations (CNVs) were identified by using the sequencing reads-based GATK4-CNV algorithm. IGHV rearrangements were obtained by using LymphoTrack® IGH FR1 Assay Panel kit. Molecular clusters were identified by an iterative algorithm that maximizes genetic distinctiveness of subgroups by reassigning patients between clusters that are created a priori based on the co-occurrence of genetic lesions. Relative survival, defined as the ratio between actuarial survival observed in the SMZL cohort and expected survival of the general population matched to patients by geographical origin, sex, age and calendar year of diagnosis, was calculated using the Ederer II method. Results. The analysis included 303 patients with a SMZL diagnosis confirmed on spleen histology. The sample size allowed to identify 30% differences in survival for molecular subgroups comprising at least 5% of cases with a statistical power between 80-100%. Median follow-up was 9.2 years. At 10 years, 85% of patients were alive, consistent with the known indolent behavior of this lymphoma. Genes recurrently affected by non-synonymous somatic mutations in >10% of SMZL included KLF2 (24%), NOTCH2 (19%), KMT2D (15%), TNFAIP3 (13%), EP300 and TP53 (10%). Deletion 7q was documented in 25% of cases and IGHV1-2*04 usage in 32%. By cluster analysis, three major molecular subgroups were identified, each of them characterized by a NOTCH pathway mutated gene (Fig. 1A). The first cluster was defined by NOTCH2 and/or KLF2 mutations and was enriched in TNFAIP3 mutations and IGHV1-2*04 gene usage (Fig. 1A). The second cluster was defined by SPEN mutations, and was enriched in KMT2D and other epigenetic gene mutations (Fig. 1A). The third cluster was enriched in NOTCH1 mutations (Fig. 1A). By relative survival analysis, the NOTCH2/KLF2 cluster showed a lower survival compared to the matched general population, indicating a significant impact of the disease on patients' expected survival (Fig. 1B). Conclusions. The large sample size and inclusion of SMZL confirmed by spleen histopathology review allowed for precise estimation of the prevalence of KLF2 and NOTCH2 mutations in this lymphoma. Three molecular clusters were identified in SMZL, each of them containing a NOTCH pathway gene, supporting the relevance of NOTCH signaling in the pathogenesis of SMZL. Patients belonging to the NOTCH2/KLF2 cluster had a lower relative survival compared to the matched general population. Disclosures Traverse-Glehen: Astra Zeneca: Other: Travel; Takeda: Research Funding. Gomes da Silva:Roche: Other: Institution's payment for consultancy, Travelling support; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees; Celgene: Other: Travelling support; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees, Institution's payment for consultancy, Travelling support; Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees; Gilead Sciences: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: lecture fees, Research Funding. Ladetto:Celgene: Honoraria; Jannsen: Honoraria; Acerta: Honoraria; Abbvie: Honoraria; Sandoz: Honoraria; Roche: Honoraria. Rambaldi:Pfizer: Consultancy; Novartis: Consultancy; Omeros: Consultancy; Italfarmaco: Consultancy; Amgen Inc.: Consultancy; Roche: Consultancy; Celgene: Consultancy. Vitolo:Takeda: Speakers Bureau; Sandoz: Speakers Bureau; Gilead: Speakers Bureau; Janssen: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau. Zinzani:MSD: Honoraria, Speakers Bureau; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; SERVIER: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; TG Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bayer: Membership on an entity's Board of Directors or advisory committees; PFIZER: Honoraria, Membership on an entity's Board of Directors or advisory committees; Merck: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Speakers Bureau; Bayer: Membership on an entity's Board of Directors or advisory committees; TG Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celltrion: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Membership on an entity's Board of Directors or advisory committees; PFIZER: Honoraria, Membership on an entity's Board of Directors or advisory committees; Verastem: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Merck: Honoraria, Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees; Astra Zeneca: Speakers Bureau. Gaidano:Roche: Consultancy, Honoraria; Gilead: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; Morphosys: Honoraria; Amgen: Consultancy, Honoraria; Janssen: Consultancy, Honoraria. Salles:Servier: Honoraria; Abbvie: Honoraria; F. Hoffmann-La Roche Ltd: Consultancy, Honoraria, Research Funding; Novartis: Consultancy, Honoraria; Amgen: Honoraria; BMS: Honoraria, Other: Advisory Board; Celgene: Honoraria, Other: Advisory Board, Research Funding; Acerta: Honoraria; Janssen: Honoraria, Other: Advisory Board; Merck: Honoraria; Pfizer: Honoraria; Morphosys: Honoraria; Gilead: Honoraria, Other: Advisory Board; Epizyme: Honoraria; Servier: Honoraria, Other: Advisory Board; Takeda: Honoraria. Zucca:Celltrion: Consultancy; AstraZeneca: Consultancy.
- Published
- 2018
16. High rate of clinical and molecular remissions in follicular lymphoma patients receiving high-dose sequential chemotherapy and autografting at diagnosis: a multicenter, prospective study by the Gruppo Italiano Trapianto Midollo Osseo (GITMO)
- Author
-
Nicola Di Renzo, Ignazio Majolino, Gino Santini, Paolo Vivaldi, Corrado Tarella, Teodoro Chisesi, Alberto De Crescenzo, Giuseppe Fioritoni, Marco Sorio, Carola Boccomini, Maurizio Martelli, Marco Ladetto, Alessio Perrotti, Maurizio Musso, Maura Brugiatelli, Sonia Vallet, P. Coser, Daniela Drandi, R Zambello, Fabio Benedetti, S. Morandi, Umberto Vitolo, Flavia Salvi, Monica Astolfi, Paolo Corradini, Andrea Gallamini, and Alessandro Pileri
- Subjects
Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,AUTOLOGOUS BONE-MARROW, NON-HODGKINS-LYMPHOMA, B-CELL LYMPHOMA, POLYMERASE-CHAIN-REACTION, MINIMAL RESIDUAL DISEASE, LOW-GRADE LYMPHOMA, TERM FOLLOW-UP, MULTIPLE-MYELOMA, BCL-2 TRANSLOCATION, INDOLENT LYMPHOMA ,Immunology ,Follicular lymphoma ,Transplantation, Autologous ,Biochemistry ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Autologous transplantation ,Prospective Studies ,Progenitor cell ,Prospective cohort study ,Lymphoma, Follicular ,Chemotherapy ,business.industry ,Remission Induction ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Debulking ,Combined Modality Therapy ,Survival Analysis ,Chemotherapy regimen ,Lymphoma ,Surgery ,Italy ,Female ,business - Abstract
Single-center experiences have shown that intensified treatments with autologous transplantation are a promising therapeutic strategy for patients with high-risk follicle-center lymphoma (FCL) at diagnosis, whereas data from prospective multicenter trials are still lacking. This paper describes the results of a prospective multicenter study of an intensified purging-free high-dose sequential (i-HDS) chemotherapy schedule with peripheral blood progenitor cell (PBPC) autografting. The main feature of this program is harvesting stem cells after intensified chemotherapeutic debulking, with no ex vivo manipulation of PBPCs. Ninety-two previously untreated patients aged 60 or younger with advanced-stage FCL were enrolled by 20 Italian centers and evaluated on an intention-to-treat basis. i-HDS proved feasible with limited toxicity (87% patients completed the planned treatment schedule). i-HDS led to a complete remission rate of 88%. The projected overall survival and disease-free survival (DFS) were, respectively, 84% and 67% at 4 years. Centralized molecular analysis showed that polymerase chain reaction-negative harvests could be collected in 47% of cases. Following autograft, 65% of molecularly evaluable patients achieved clinical and molecular remission. The projected DFS at 4 years of this subgroup is 85%. This result emphasizes the importance of achieving maximal tumor reduction in these patients. In conclusion, our data show that highly effective intensified treatments can now be routinely offered to young patients with poor-risk FCL even at small institutions, with no need for sophisticated and expensive cell manipulation procedures.
- Published
- 2002
17. Rituximab, Bendamustine and Cytarabine (RBAC500) As Induction Therapy in Elderly Patients with Mantle Cell Lymphoma: Final Results of a Phase 2 Study from the Fondazione Italiana Linfomi
- Author
-
Carlo Visco, Giovannino Ciccone, Alice Di Rocco, Simone Ferrero, Alberto Fabbri, Andrea Evangelista, Marco Ruggeri, Angelo Michele Carella, Stefano Pileri, Luigi Rigacci, Monica Tani, Manuel Gotti, Marco Ladetto, Daniela Barbero, Graziella Pinotti, Gianluca Gaidano, Caterina Patti, Silvia Finotto, Michele Spina, Maurizio Martelli, Anna Lia Molinari, Renato Zambello, Umberto Vitolo, Luca Nassi, Flavia Salvi, and Annalisa Chiappella
- Subjects
Bendamustine ,medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Surgery ,03 medical and health sciences ,Regimen ,0302 clinical medicine ,International Prognostic Index ,Maintenance therapy ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Mantle cell lymphoma ,business ,Febrile neutropenia ,Progressive disease ,030215 immunology ,medicine.drug - Abstract
Background: The combination of rituximab (R, 375 mg/m2 intravenously [IV], day 1), bendamustine (B, 70 mg/m2IV, days 2 and 3), and cytarabine (800 mg/m2, IV on days 2 to 4) was highly active in patients with mantle-cell lymphoma (MCL) in a phase 2 study [R-BAC; Visco et al, JCO 2013]. This regimen was well tolerated, but hematologic toxicity was quite relevant, especially in terms of transient grade 3 to 4 thrombocytopenia (76% of cycles). Aiming at reducing hematologic toxicity, the Fondazione Italiana Linfomi (FIL) designed a phase 2 trial adopting the R-BAC schedule, but lowering cytarabine dose to 500 mg/m2 (RBAC500). Materials and Methods: Patients with newly diagnosed MCL, aged 61 to 80 years, not eligible for autologous transplant and fit according to the comprehensive geriatric assessment, were enrolled. Patients presenting with non-nodal leukemic disease were excluded. The primary endpoints were complete remission rate (CR) measured by FDG-PET according to Cheson criteria 2007, and safety. Secondary endpoints included rate of molecular response (MR) by nested-PCR using patient specific IGH or BCL1 based targets, progression-free (PFS) and overall survival (OS). The study was conducted according to the Bryant and Day two-stage design. Results: From May 2012 to February 2014, 57 patients with MCL from 29 centers were recruited and treated. Central pathology revision was performed in 87% of cases. Median age was 71 years (range 61-79), 75% were males, and 91% had Ann Arbor stage III/IV disease. Mantle Cell International Prognostic Index (MIPI) was low in 15%, intermediate in 40%, high in 45%, Ki-67 was ≥30% in 31%, and 9% had the blastoid cytological variant. Overall, 53 patients (91%) received at least 4 cycles, while 36 (63%) had 6 cycles (median 5.3 cycles per patient). Fifteen patients (26%) discontinued treatment before reaching cycle 6 because of toxicity/adverse events, that mainly consisted of prolonged hemato-toxicity between cycles. Only one patient discontinued due to progressive disease. Grade 3 or 4 neutropenia and thrombocytopenia were observed in 49% and 52% of administered cycles, respectively. Febrile neutropenia occurred in 6% of cycles. Extra-hematologic toxicity was mainly cardiac (5%). Overall response rate was 96%, and CR was 93%. The MR rate at the end of treatment was 76% on peripheral blood and 55% on bone marrow (BM) samples. With a median follow-up of 34 months (28-52), the 2-years PFS (± confidence interval) was 81%±5% and the OS 85%±4%. Elevated Ki-67 (≥30%), and the blastoid variant were the strongest independent predictors of adverse PFS. Patients with either of these two features (33%), had a significantly inferior PFS (41% vs 97% after 34 months) compared to patients with classical/pleomorphic variants and low proliferative index (p Conclusions: The R-BAC500 regimen can be safely administered as first line therapy to elderly patients with MCL. Hematologic toxicity is substantially reduced compared to our previous experience. With 93% of FDG-PET negative CR, and a 2-years PFS of 81% without maintenance therapy, the R-BAC500 regimen is a highly effective treatment for patients with MCL, and compares favourably with previously reported regimens in this patient population, including R-bendamustine. Figure 1 Figure 1. Disclosures Visco: Gilead: Speakers Bureau; Lundbeck: Consultancy; Mundipharma: Research Funding; Celgene: Speakers Bureau. Spina:Mundipharma: Membership on an entity's Board of Directors or advisory committees, Other: Speaker Fee; Teva Pharmaceuticals Industries: Membership on an entity's Board of Directors or advisory committees, Other: Speaker Fee. Di Rocco:Celgene: Honoraria. Carella:Millenium: Speakers Bureau; Genentech: Speakers Bureau. Vitolo:Roche: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria; Gilead: Honoraria; Celgene: Honoraria.
- Published
- 2016
18. Identification of DNA Copy Number Variations Associated with the Clinical Outcome in Young Mantle Cell Lymphoma Patients Treated with Cytarabine-Based High Dose Sequential Chemotherapy and Autologous Stem Cell Transplantation in the Prospective from the MCL0208 Phase III Trial from Fondazione Italiana Linfomi (FIL)
- Author
-
Marco Ladetto, Caterina Stelitano, Francesco Bertoni, Elisa Doni, Simone Ferrero, Alessandra Flavia Salvi, Sergio Cortelazzo, Michael Mian, Fabio Benedetti, Valeria Spina, Davide Rossi, Andrea Rinaldi, Fary Diop, Filippo Gherlinzoni, Alessio Bruscaggin, Gianluca Gaidano, Filippo Ballerini, and Ivo Kwee
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Immunology ,Aggressive lymphoma ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Autologous stem-cell transplantation ,Internal medicine ,Medicine ,Progression-free survival ,Lenalidomide ,Univariate analysis ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,Chemotherapy regimen ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cytarabine ,Mantle cell lymphoma ,business ,medicine.drug - Abstract
Background. Mantle Cell Lymphoma (MCL)represents an aggressive lymphoma for which an effective treatment has still to be determined. The FIL-MCL0208 phase III trial (EudraCTNumber: 2009-012807-25) is exploring R-CHOP followed byi) high-dose cytarabine, autologous stem cell transplantation, and ii) randomization between lenalidomide maintenance vs observation (Cortelazzo et al, EHA 2015). We performed genome-wide DNA profiling to identify unbalanced copy number variations (CNVs) with a clinical significance in patients enrolled in the FIL-MCL0208 phase III trial. Patients and Methods. The study included untreated, advanced stage MCL patients ( Results. 161 patients were currently evaluable for CNVs and clinical outcome. Patients had an intermediate/high-risk MIPI and a Ki67 ³ 30% in 43% and 42% of the cases, respectively. Twenty-five recurrent unbalanced CNVs were defined (Table 1). By multiple test corrected univariate analysis, seven CNVs had a negative impact on PFS: +3, 7p gain, 9p loss (CDKN2A), 17p loss (TP53), 8p loss, and 2 losses at 22q (Table 1). MIPI (intermediate/high vs low risk), Ki67+ and the TP53/KMT2D model (Rossi et al, ASH 2015) were also statistically significant. Combining CNVs and mutations, TP53 was inactivated in 27/147 cases (18%): mutated/deleted in 7/147 (5%), deleted but not mutated in 14/147 (10%), and mutated but not deleted in 6/147 (4%). The negative prognostic impact was equal for all the 3 inactivation modalities, which were then considered as a single group for further analyses. ATM was inactive in 69/147 (47%): mutated/deleted in 24/147 (16.3%), deleted in 12/147 (8%), and mutated in 33/147 (22.4%). KMT2D (MLL2) was inactive in 18/147 (12%): mutated/deleted in 1/147 ( The lesions with a significant impact at univariate were included in a multivariate analysis alongside MIPI, KMT2D inactivation and Ki67+ as continuous variable. Only TP53 inactivation by deletion and/or mutation, 7p22.2-p12.1 gain and KMT2D inactivation maintained their independent prognostic significance. Conclusions. Genome wide DNA profiling in the FIL-MCL0208 phase III trial identified lesions, namely TP53 and KMT2D inactivation and gains at 7p, which maintain a poor outcome significance in young MCL patients even following high-dose cytarabine and autologous stem cell transplantation. Disclosures Rossi: Gilead: Honoraria, Research Funding; Abbvie: Honoraria; Janseen: Honoraria. Stelitano:Azienda Ospedaliera: Employment. Gaidano:Janssen: Consultancy, Honoraria, Speakers Bureau; Novartis: Consultancy, Honoraria, Speakers Bureau; Gilead: Consultancy, Honoraria, Speakers Bureau; Morphosys: Consultancy, Honoraria; Roche: Consultancy, Honoraria, Speakers Bureau; Karyopharm: Consultancy, Honoraria.
- Published
- 2016
19. Outcomes for Elderly Patients (pts) with Follicular Lymphoma (FL) Using Individual Patient Data (IPD) from 5922 Pts in 18 Randomized Controlled Trials (RCTs): a Follicular Lymphoma Analysis of Surrogate Hypothesis (FLASH) Group Study
- Author
-
Mathias J. Rummel, Pauline Brice, Bruce A. Peterson, Eva Kimby, Wolfgang Hiddemann, Marco Ladetto, Gilles Salles, Michael Herold, Qian Shi, Robert Marcus, Daniel J. Sargent, Tina Nielsen, Anton Hagenbeek, Fang-Shu Ou, Umberto Vitolo, Eva Hoster, Christopher R. Flowers, Howard S. Hochster, Sabine De Bedout, and Franck Morschhauser
- Subjects
medicine.medical_specialty ,Group study ,business.industry ,Time to progression ,Immunology ,Early disease ,Disease progression ,Follicular lymphoma ,Cell Biology ,Hematology ,Patient data ,medicine.disease ,Biochemistry ,law.invention ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,030220 oncology & carcinogenesis ,Family medicine ,Medicine ,business ,030215 immunology - Abstract
Background: Limited data exist to describe the clinical features and outcomes for elderly pts with FL. The FLASH group performed a prospectively planned pooled analysis of IPD from first-line RCTs and examined associations between age ( 70 years), clinical characteristics and FL outcomes. Methods: We identified 18 randomized controlled multicenter clinical trials in the FLASH database which enrolled elderly pts (> 70 yrs). From these 18 studies, 5922 previously untreated FL pts were included for this analysis. Complete response (CR) at 24 and 30 months were defined as whether the pt achieved and remains in CR at 24 months and 30 months after initiation of induction treatment. PFS24 is defined as proportion of pts progression-free and alive at 24 months post-randomization. Time to progression (TTP) and overall survival (OS) were defined as time from randomization to the date of progression (censoring for death without progression) and date of death due to any cause, respectively. Progression-free survival (PFS) was defined as time from randomization to the date of progression or death, due to any cause, whichever comes first. Non-lymphoma death was defined as death without prior progression. Early disease outcomes, i.e. CR24, CR30, and PFS24 were primary outcomes; secondary outcomes were TTP, OS, and PFS. For time-to-event outcomes, Kaplan-Meier method and log-rank test were used for univariate estimation and comparison; stratified Cox proportional hazard modeling was used for multivariable analyses. Cumulative incidence methods were used to model PFS while treating disease progression as the primary event of interest and non-lymphoma death as a competing risk. For binary outcomes, multivariable logistic regression was used to estimate the association between age groups and outcomes. Variable adjusted in the regression models are FLIPI score without the age component, ECOG performance status (>= 2 vs < 2), and rituximab use. Results: Among 5922 previously untreated FL pts from 18 RCTs, 63% (n = 3728) were 70 yrs. Pts age > 70 (vs. = 2 (8.8% vs 5.0%, p = 0.0004), and elevated β2-microglobulin (68% vs 49%, p < 0.001), less often had > 4 lymph nodes involved (54% vs 65%, p < 0.001), and had similar FLIPI scores without age component (p = 0.17). There were no significant differences between groups in: Ann Arbor stage (94% vs 95% stage III/IV) or rituximab use (62% vs 58%). Median survival follow-up was 5.6 yrs. Pts > 70 yrs did not differ from pts 70 and 70 and 70 and 70 with no difference in disease progression (Figure). In regression models, adjusting for rituximab use, ECOG PS >= 2, and FLIPI score without the age component, age > 70 was a significant predictor of OS and PFS (due to higher incidence of non-lymphoma death), but not PFS24, CR24 or CR30 (Table). Conclusions: FL pts > 70 yrs treated on trials have similar early disease outcomes to younger patients. There is no disease-specific outcome difference between age groups. Age alone should not disqualify patients from standard treatments or RCTs. Disclosures Flowers: Genentech: Consultancy, Research Funding; NIH: Research Funding; Mayo Clinic: Research Funding; TG Therapeutics: Research Funding; Millenium/Takeda: Research Funding; ECOG: Research Funding; Acerta: Research Funding; Pharmacyclics, LLC, an AbbVie Company: Research Funding; Roche: Consultancy, Research Funding; AbbVie: Research Funding; Infinity: Research Funding; Gilead: Consultancy, Research Funding. Ou:Mayo Clinic: Employment. Shi:Mayo Clinic: Employment. Hochster:Genentech: Consultancy, Other: Consultancy fees for advisory boards for GI cancer and bevacizumab with amounts within Yale University de minims guidelines. Brice:Roche: Honoraria; Takeda Pharmaceuticals International Co.: Honoraria, Research Funding; Bristol Myers-Squibb: Honoraria; Gilead: Honoraria; Seattle Genetics: Research Funding. Hiddemann:Roche: Other: Grants; Roche: Membership on an entity's Board of Directors or advisory committees; Genentech: Other: Grants. Marcus:Takeda: Consultancy, Speakers Bureau; Roche: Consultancy, Speakers Bureau. Kimby:Abbvie: Consultancy, Honoraria; Jansen: Consultancy, Honoraria; Celgene: Honoraria; Baxalta: Consultancy; Gilead: Honoraria. Herold:Gilead: Other: Personal fees from member advisory board; Celgene: Honoraria; Genentech: Other: Grants; Roche: Honoraria, Other: Grants. De Bedout:Celgene: Employment. Nielsen:Hoffmann-La Roche: Employment. Morschhauser:Roche: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Servier: Consultancy, Honoraria; Gilead Sciences: Consultancy, Honoraria; Janssen: Honoraria. Rummel:Mundipharma GmbH: Other: Personal fees, Research Funding; Roche Pharma AG: Other: Personal fees, Research Funding. Vitolo:Gilead: Other: Honoraria for lectures; Takeda: Other: Honoraria for lectures; Roche: Membership on an entity's Board of Directors or advisory committees, Other: Honoraria for lectures; Janssen: Membership on an entity's Board of Directors or advisory committees, Other: Honoraria for lectures. Salles:Celgene: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Gilead: Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Roche/Genentech: Consultancy, Honoraria, Research Funding; Mundipharma: Honoraria. Sargent:Celgene: Research Funding.
- Published
- 2016
20. Distinct Immunogenetic Signatures in IgA Versus IgG Multiple Myeloma
- Author
-
Simone Ferrero, Theodoros Moysiadis, Evangelos Terpos, Ramón García Sanz, Andreas Agathangelidis, Chrysoula Belessi, Evdoxia Hatjiharissi, Chrysavgi Lalayanni, Alejandro Medina, Elisa Genuardi, Kostas Stamatopoulos, Apostolia Papalexandri, Achilles Anagnostopoulos, Anastasia Hadzidimitriou, Katerina Gemenetzi, Maria Papaioannou, Marco Ladetto, and Cristina Jimenez
- Subjects
0301 basic medicine ,Immunoglobulin A ,biology ,business.industry ,Immunology ,Somatic hypermutation ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Isotype ,Immunoglobulin G ,03 medical and health sciences ,030104 developmental biology ,Immunoglobulin class switching ,biology.protein ,medicine ,Antibody ,IGHV@ ,business ,Multiple myeloma - Abstract
Immunogenetic analysis of MM has proven instrumental in elucidating disease ontogeny e.g. by revealing the clonal relationship between switch variants expressed by the bone marrow plasma cells and myeloma progenitors in the marrow and blood; demonstrating the marked under-representation of the inherently autoreactive IGHV4-34 gene; and, identifying patterns of somatic hypermutation (SHM) indicative of post-germinal center derivation. Yet, limited information exists about the composition of the immunoglobulin (IG) gene repertoire in MM cases expressing different heavy chain isotype, in particular A versus G. This is relevant in light of studies showing an overall higher SHM impact in CD27+IgA+ compared to CD27+IgG+ normal memory B cells, perhaps reflecting a distinct location of the immune response, especially considering that IgA class switching mostly occurs in mucosa-associated lymphoid tissues. From a clinical perspective, it is also relevant to note that IgA patients exhibit a higher incidence of the t(4;14) translocation, shorter progression-free survival and worse median overall survival compared to IgG patients. Here, we explored potential differences in the immunoprofiles of IgA versus IgG MM focusing on IG gene repertoire and SHM characteristics. In total, 428 patients with a diagnosis of MM following the IMWG criteria from collaborating institutions in Greece, Italy and Spain (n=355) or retrieved from the LIGM-DB (n=73) were included in the study. Of these, 135 and 293 belonged to IgA and IgG MM groups, respectively. Amongst the evaluated productive IG rearrangements, IGHV3 subgroup genes predominated in both groups (IgA: 58.5%; IgG: 52.2%). However, at the individual gene level, major asymmetries were noted, since only 7 IGHV genes accounted for 41.6% of the IgA and 46.7% of the IgG cases, respectively. Of these, 3 genes were shared between IgA and IgG MM cases: IGHV3-30 (IgA: 11.9% - IgG: 13.3%), IGHV3-23 (IgA: 5.2% - IgG: 6.8%) and IGHV3-9 (IgA: 6.7% - IgG: 4.4%), whereas the remaining 4 of the 7 most frequent genes were specific for each group with significant (p Disclosures Terpos: Celgene: Honoraria; Novartis: Honoraria; Genesis: Consultancy, Honoraria, Other: Travel expenses; BMS: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Other: Travel expenses, Research Funding; Takeda: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Other: Travel expenses, Research Funding. Stamatopoulos:Gilead: Consultancy, Honoraria, Research Funding; Abbvie: Honoraria, Other: Travel expenses; Novartis: Honoraria, Research Funding; Janssen: Honoraria, Other: Travel expenses, Research Funding.
- Published
- 2016
21. LONG-TERM Outcome of a Fondazione Italiana Linfomi Study Comparing Short Rituximab Maintenance Vs Observation after Brief First-LINE R-FND Chemoimmunotherapy Followed By Rituximab Consolidation in Elderly Patients with Advanced Follicular Lymphoma (FL)
- Author
-
Annarita Conconi, Annalisa Chiarenza, Giuseppe Rossi, Francesca Dutto, Stefano Volpetti, Benedetta Puccini, Eleonora Russo, Stefan Hohaus, Simone Ferrero, Chiara Bottelli, Chiara Rusconi, Luca Baldini, Umberto Vitolo, Federico De Angelis, Carola Boccomini, Marco Ladetto, Claudia Castellino, Stefano Sacchi, Francesco Merli, and Andrea Evangelista
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Immunology ,Population ,Follicular lymphoma ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Fludarabine ,Surgery ,Regimen ,Chemoimmunotherapy ,Internal medicine ,medicine ,Rituximab ,Progression-free survival ,business ,education ,medicine.drug - Abstract
Introduction: we previously reported (Vitolo U, JCO 2013) the results of a randomized study with brief first-line chemoimmunotherapy followed by rituximab maintenance vs observation. With a median follow-up of 42 months, 3-year Progression Free Survival (PFS) and Overall Survival (OS) were 66% and 89%, respectively. The addition of Rituximab maintenance gave a benefit to the patients: 2-year PFS was 81% for rituximab maintenance versus 69% for observation with a HR of 0.63 (95% CI: 0.38-1.05, p=0.079), although not statistically significant. Moreover we also found that achievement of Minimal Residual Disease (MRD) negativity predicted a better PFS: 3-year PFS 72% vs 39%, HR 3.1 (Ladetto M, Blood 2013). Overall these data showed the good efficacy of this brief chemoimmunotherapy regimen in elderly FL patients. Aim of this analysis was to report long-term outcome and long-term toxicities of this regimen. Methods: From January 2004 to December 2007, 242 treatment-naive patients aged 60-75 years with FL Grade I, II and IIIa were enrolled by 33 FIL centres. Patients had to have advanced (high tumor burden stage II or stage III-IV) disease requiring treatment: 4 monthly courses of R-FND (standard doses of Rituximab, Fludarabine, Mitoxantrone, Dexamethasone) every 28 days followed by 4 weekly Rituximab infusions as consolidation. Responders patients [complete remission (CR) + unconfirmed CR + partial remission (PR)] were randomized to brief rituximab maintenance (Arm A), once every 2 months for a total of 4 doses, or observation (Arm B). MRD for the bcl-2/IgH translocation was determined on bone marrow cells in a centralized laboratory belonging to Euro-MRD consortium, using qualitative and quantitative PCR. Results: a total of 234 patients began chemoimmunotherapy: after induction and consolidation treatment overall response rate was 86%, with 69% CR. Of these, 210 completed the planned treatment and 202 responders were randomized. Up to date, median follow-up were 96 months from enrollment and 87 months from randomization; additional follow-up data were available for 127/146 (87%) not relapsed/progressed patients. Five- and 7-year PFS for the whole population were 57% and 51%, respectively; 5- and 7-year OS for the whole population were 85% and 80%, respectively. From enrollment, an advantage in term of PFS and also OS was observed in FLIPI low risk patients: 7-year PFS was 67% for low risk versus 38% for intermediate-high risk patients (p Conclusions: the present long-term results of this trial with a prolonged follow-up of 7 years confirm that a good outcome is achievable in elderly FL patients with a short-term chemoimmunotherapy (R-FND + Rituximab consolidation) with a 7-year PFS of 51% and low toxicity. In addition these results did not show clear evidence in favor of a shortened Rituximab maintenance after R-fludarabine containing chemotherapy. Conversely, the achievement of PCR negativity maintains predictive value for a better outcome. Figure 1. Figure 1. Disclosures Off Label Use: Rituximab maintenance was not licensed in first-line treatment for follicular lymphoma at that time in Italy; Rituximab was provided free by Roche.
- Published
- 2015
22. Highly Sensitive Droplet Digital PCR for MYD88L265P Mutation Detection and Minimal Residual Disease Monitoring in Waldenström Macroglobulinemia
- Author
-
Luigia Monitillo, Federica Cavallo, Mario Boccadoro, Elisa Genuardi, Marika Vasta, Giulia Verardo, Eleonora Marzanati, Simone Ferrero, Barbara Mantoan, Paola Ghione, Daniela Drandi, Marina Ruggeri, Daniela Barbero, Marco Ladetto, Paola Omedè, and Daniele Grimaldi
- Subjects
Oncology ,Sanger sequencing ,medicine.medical_specialty ,business.industry ,Immunology ,Waldenstrom macroglobulinemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Minimal residual disease ,Lymphoplasmacytic Lymphoma ,symbols.namesake ,Circulating tumor cell ,Real-time polymerase chain reaction ,Internal medicine ,medicine ,symbols ,Digital polymerase chain reaction ,business ,Multiple myeloma - Abstract
Background. Recently, the somatic MYD88L265P mutation has been found as the hallmark of Waldenström Macroglobulinemia (WM), being detectable in nearly 90% of cases, as well as in up to 50% of IgM MGUS, rarely in other non-Hodgkin lymphomas and never in multiple myeloma (MM). Beyond its potential diagnostic role, this mutation has been associated with tumor growth and therapy resistance. Moreover, MYD88L265P might represent an ideal marker for minimal residual disease (MRD) monitoring in a disease whose therapeutic scenario has been rapidly changing, with many new available and highly effective drugs (nucleoside analogues, proteasome and BTK-inhibitors). However, the current MYD88L265P allele-specific quantitative PCR (ASqPCR) diagnostic tool lacks sensitivity (1.00E-03) and thus is not suitable for MRD. Moreover, is not useful to test peripheral blood (PB), that harbors low concentrations of circulating tumor cells (especially after immunochemotherapy), neither to assess cell-free DNA (cfDNA), usually present at very low amount in plasma. Therefore, our study aims: 1) to assess whether a highly sensitive tool as droplet digital PCR (ddPCR) might be helpful in MYD88L265P screening; 2) to evaluate whether MYD88L265P might be a suitable marker for MRD monitoring in WM. Methods. Bone marrow (BM) and PB samples were collected at diagnosis and during follow-up from a local series of patients affected by WM, IgM MGUS and IgG-secreting lymphoplasmacytic lymphoma (LPL), as well as samples from healthy subjects and MM were used as negative controls. Genomic (gDNA) and cell-free DNA (cfDNA) were extracted as recommended (Qiagen). MYD88L265P was assessed on 100 ng of gDNA by ASqPCR as previously described [Xu 2013] and by ddPCR, using a custom dual labelled probe assay (Bio-Rad). When available, 50 ng of cfDNA were tested for MYD88L265P, only by ddPCR. ddPCR was performed on 20 µl of reaction at 55°C for 40 cycles, run on QX100 droplet reader and analyzed by QuantaSoft v1.6.6 (Bio-Rad). MYD88L265P ASqPCR level was estimated as described [Treon 2012]. ΔCT Results. Once the ddPCR assay was optimized, the sensitivity of MYD88L265P ddPCR was compared to ASqPCR on a ten-fold serial dilution standard curves built with a 70% MYD88L265P mutated WM sample, previously identified by Sanger sequencing [Treon 2012]. Whereas ASqPCR confirmed the reported sensitivity of 1.00E−03, ddPCR reached a sensitivity of 5.00E−05. Thereafter, overall 105 samples (48 BM, 57 PB, 52 diagnosis and 53 follow up) from 58 patients (49 WM, 5 IgM MGUS and 4 LPL) as well as 20 controls (15 healthy subjects and 5 MM) were tested by both methods. 32/33 (97%) diagnostic BM scored positive for MYD88L265P by both ddPCR and ASqPCR (being the only one negative a WM), while ddPCR, was able to detect more mutated cases, than ASqPCR, among diagnostic PB samples: 15/19 (79%) vs 9/19 (47%) (Table1). Moreover, to investigate whether the MYD88L265P ddPCR tool could be used for MRD detection we compared it to the standardized IGH-based MRD. An IGH-based MRD marker was found in 40/53 (75%) patients (37 WM and 3 LPL). Five Patients, so far analyzed, with baseline and follow up samples (18 BM, 5 PB) showed highly superimposable results between the two methods. Finally, pivotal results on cfDNA from 10 patients showed higher median levels of MYD88L265P mutation in plasma if compared to PB. Conclusions. We developed a new tool for diagnosis and MRD monitoring in WM, showing that: 1) ddPCR is a highly sensitive tool for MYD88L265P detection, especially useful in low infiltrated samples, like PB; 2) MYD88L265P can be effectively and easily used for MRD monitoring in WM, achieving similar results to standardized IGH-based MRD; 3) cfDNA recovered from plasma might be an attractive alternative for MYD88L265P detection, deserving further investigation. Methodological validation against IgH-based MRD detection and Flow cytometry and correlations with clinical impact are currently ongoing on external samples series. Table 1.PATIENTSWM (45)LPL (2)IgM MGUS (5)TISSUEBMPBBMPBBMPBSAMPLES31141114MYD88L265P ddPCR/ASqPCR30/3011/71/10/01/14/2 TABLE 1. MYD88L265P mutation detection in diagnostic samples: ddPCR vs ASqPCR Disclosures Boccadoro: Sanofi: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees; Onyx Pharmaceuticals: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen-Cilag: Consultancy, Membership on an entity's Board of Directors or advisory committees.
- Published
- 2015
23. A Molecular Model for the Prediction of Progression Free Survival in Young Mantle Cell Lymphoma Patients Treated with Cytarabine-Based High Dose Sequential Chemotherapy and Autologous Stem Cell Transplantation: Results from the MCL0208 Phase III Trial from Fondazione Italiana Linfomi (FIL)
- Author
-
A. L. Molinari, Luigia Monitillo, Maria Gomes da Silva, Valeria Spina, Davide Rossi, Gianluca Gaidano, Armando Santoro, Daniela Barbero, Andrés J.M. Ferreri, Simone Ferrero, Paola Ghione, Alice Di Rocco, Giovannino Ciccone, Sergio Cortelazzo, Vittorio Stefoni, Marco Ladetto, and Alessio Bruscaggin
- Subjects
Oncology ,medicine.medical_specialty ,Mutation ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease_cause ,Interim analysis ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Autologous stem-cell transplantation ,Internal medicine ,Clinical endpoint ,medicine ,Cytarabine ,Mantle cell lymphoma ,Progression-free survival ,business ,medicine.drug - Abstract
Background. Recent studies have described the landscape of recurrently mutated genes in mantle cell lymphoma (MCL), including genes involved in DNA damage response/cell cycle (ATM, TP53, CCND1), epigenetic regulation (KMT2D also known as MLL2, WHSC1), and cell signaling (BIRC3, TRAF2, NOTCH1). However, with the exception of TP53 abnormalities, little is known about the clinical relevance of recurrent mutations in MCL. Thus, we performed deep sequencing analysis of a MCL gene panel in the prospective series of patients enrolled in the ongoing FIL-MCL0208 phase III trial (EudraCTNumber: 2009-012807-25). Patients and Methods. The study included untreated, advanced stage 70% of cases. The gene panel was analyzed in tumor DNA from baseline bone marrow CD19+ purified MCL cells and, for comparative purposes to filter out polymorphisms, in the paired normal genomic DNA (available in 55% of cases) using a TruSeq Custom Amplicon target enrichment system followed by deep next generation sequencing (Illumina, median depth of coverage 2356x). Variants represented in >10% of the alleles were called with VarScan2 with the somatic function when the paired germline DNA was available. For patients lacking germline DNA, a bioinformatic pipeline including a number of stringent filters was applied to protect against the misclassification of polymorphisms as somatic variants. Primary endpoint of the analysis was progression free survival (PFS). Results. Out of the enrolled patients, 151 are currently evaluable for mutations and clinical outcome (median age: 57 years, range 35-66; males 75%). Among prognostic factors, the MIPI was intermediate or high-risk in 49% of patients, the Ki67 ≥30% in 39%, and blastoid histology occurred in 8%. At the first planned interim analysis, median follow-up of alive patients was 26 months. At 2-years, 79% of patients were progression free and 91% alive (Cortelazzo et al EHA 2015). Overall, at least one mutation was detected in 106/151 cases (70%), including mutations of ATM in 42% of cases, CCND1 in 14%, WHSC1 in 13%, KMT2D in 12%, TP53 in 7%, NOTCH1 in 6%, BIRC3 in 5% and TRAF2 in 1% (Figure 1A). By univariate analysis, mutations of TP53 (2-years PFS 48% vs 82%; p Conclusions. Though limited by the short follow-up, our data show that: i) the combination of two genetic biomarkers (i.e. TP53 and KMT2D mutations) allows to predict the benefit that young MCL patients can gain from a cytarabine-based high dose sequential chemotherapy followed by autologous stem cell transplantation; ii) intensive chemotherapy does not overcome the negative prognostic impact of TP53 mutations; and iii) KMT2D mutations may represent a novel genetic biomarker in MCL patients. Figure 1. Figure 1. Disclosures Santoro: Celgene: Research Funding.
- Published
- 2015
24. Phase II Study of the Fondazione Italiana Linfomi on Gemcitabine Plus Romidepsin (GEMRO Regimen) in Relapsed and Refractory Peripheral T-Cell Lymphoma Patients
- Author
-
Letizia Gandolfi, Pier Luigi Zinzani, Alessandro Broccoli, Paolo Corradini, Vittorio Stefoni, Lucia Farina, Enrico Derenzini, Annalisa Chiappella, Lorella Orsucci, Francesco Spina, Federico Monaco, Cinzia Pellegrini, Flavia Salvi, Lorenzo Tonialini, Umberto Vitolo, Anna Dodero, Marco Ladetto, Lisa Argnani, Federica Quirini, and Beatrice Casadei
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Phases of clinical research ,Salvage therapy ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Gemcitabine ,Surgery ,Romidepsin ,Regimen ,Internal medicine ,Clinical endpoint ,Medicine ,business ,medicine.drug - Abstract
Introduction. Relapsed and primary refractory peripheral T-cell lymphomas (PTCL) show a dismal outcome, with a 5-year overall survival of only 30%. There is no standard salvage chemotherapy for these patients. Gemcitabine was proved to be an effective monotherapy, yelding 60-70% overall response rates in patients with advanced heavily pre-treated disease. Romidepsin, a histone deacetylase inhibitor recently approved by Food and Drug Administration, has demonstrate an overall response rate (ORR) of 30% and a complete response (CR) rate of 16%. We have recently designed a multicentric trial to investigate the role of the combination of gemcitabine plus romidepsin (GEMRO regimen) in relapsed or refractory PTCL, looking for a potential synergistic effect of the two drugs. Methods. Twenty relapsed/refractory PTCL patients were included in a multicentric, prospective phase II trial which contemplated an induction with romidepsin 12 mg/m2 intravenously (i.v.) on days 1, 8, 15, and gemcitabine, 800 mg/m2 i.v. on day 1 and 15, for 6 cycles, each cycle to be repeated every 28 days. After the induction phase, patient who obtained at least a partial remission (PR) proceeded onto romidepsin maintenance at the dose of 14 mg/m2 i.v. until disease progression. The primary endpoint was to evaluate the efficacy of GEMRO regimen after the induction phase, as assessed by complete response (CR) rate; safety assessment was regarded as a secondary objective. The trial was registered under EudraCT (2012-001404-38). Results. Twenty patients have been recruited for this study. At present time, all patients underwent the induction phase and are evaluable for response and toxicity. The median age of patients was 55 years (range, 24-77). According to histology, 10 patients had PTCL not otherwise specified, 9 had an angioimmunoblastic T-cell lymphoma, 1 had a kinase negative anaplastic large cell lymphoma. The median number of prior therapies was 2 (range, 1-4); 7/20 (35%) patients had failed a prior stem cell transplant. Nineteen out of 20 (95%) patients presented with advanced stage. At the end of induction phase, the ORR was 31% including 2 CRs and 3 PRs. One of the 2 CR patients discontinued the treatment after 4 cycles due to cardiac toxicity, however maintaining a continuous CR with a follow up of 2 years. The other CR patient is still on treatment in maintenance phase. Grade ≥3 adverse events were represented by thrombocytopenia (60%), neutropenia (50%), and anemia (20%). Conclusions. To date, data failed to show a superiority of the GEMRO combination regimen over single agent romidepsin as salvage therapy for refractory or relapsed PTCL patients. More mature data and an adequate follow-up will be required to better understand the role of this combination regimen. Disclosures Zinzani: Takeda: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Gilead: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Pfizer: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; J&J: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Celgene: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau.
- Published
- 2015
25. Identification of a Novel Gene Expression Signature in Mantle Cell Lymphoma from the Fondazione Italiana Linfomi (FIL)-MCL-0208 Trial: A Focus on the B Cell Receptor Pathway
- Author
-
Alberto Zamò, Stefano Luminari, Paola Omedè, Tiziana D'Agaro, Marco Ladetto, Michele Dal Bo, Umberto Vitolo, Sergio Cortelazzo, Andrea Evangelista, Alessandro Re, Simone Ferrero, Riccardo Bomben, Chiara Rusconi, Angelo Michele Carella, Valter Gattei, Luigi Rigacci, and Luca Arcaini
- Subjects
Genetics ,Immunology ,breakpoint cluster region ,Cell Biology ,Hematology ,Gene signature ,Biology ,CD79B ,medicine.disease ,Biochemistry ,Gene expression profiling ,Transplantation ,hemic and lymphatic diseases ,Cancer research ,medicine ,Autologous transplantation ,Mantle cell lymphoma ,IGHV@ - Abstract
Background. The aggressive clinical behavior of mantle cell lymphoma (MCL) is attributed to specific genetic and molecular mechanisms involved in its pathogenesis, mainly the t(11;14)(q13;q32) traslocation and cyclin D1 (CCND1) overexpression. Nevertheless, evidence of a certain degree of clinical/biological heterogeneity has been disclosed by gene expression profile (GEP) and (immuno)genetic/immunohistochemistry studies. Aim. To use a GEP approach to identify MCL subsets with peculiar clinical/biological features in the context of MCL patients treated homogeneously with an autologous transplantation-based program. Methods. The study was based on a cohort of 42 MCL cases enrolled in the Fondazione Italiana Linfomi (FIL)-MCL-0208 randomized Italian clinical trial. Purified clonal CD19+ MCL cells were obtained by high-speed cell sorting of peripheral blood MCL samples. GEP experiments were performed in 30 cases, with Agilent platform. Bioinformatics analyses were performed by Gene Springs and Gene Set Enrichment Analysis (GSEA) software. Gene signature validations were performed by quantitative real time PCR (QRT-PCR). Results. i)Unsupervised and supervised analyses. Unsupervised analysis by principal component analysis (PCA) was able to divide the cohort in two main subgroups named PCA1 (12 cases) and PCA2 (18 cases). Supervised analysis by segregating cases according to the PCA1 and PCA2 classification defined a gene expression signature of 710 gene (234 up-regulated) that highlighted a constitutive overexpression of genes of the BCR signaling pathway. Consistently,GSEA showed a significant enrichment of genes belonging to 3 gene sets related to BCR signaling. ii) Identification of a "PCA2-type" gene signature. By merging the list of differentially expressed genes according to supervised analysis of GEP data and the gene list related to BCR signaling according to GSEA, a group of 9 genes, all overexpressed in PCA2 cases, i.e. AKT3, BLNK, BTK, CD79B, PIK3CD, SYK, BCL2, CD72, FCGR2B, was obtained. Among these genes, a subgroup of 6 genes, i.e. AKT3, BLNK, BTK, CD79B, PIK3CD, SYK, was selected for the direct involvement in the BCR pathway, and utilized for further validations. iii) Generation of a 6-gene prediction model. The selected 6 genes were then utilized to generate a prediction model by using 20 cases as training sub-cohort and the remaining 10 cases as validation cohort. By this approach, 9/10 cases of the validation cohort were correctly assigned according to the PCA2/PCA1 classification. The model was re-tested by QRT-PCR in 24 cases used in the GEP (16 for training and 8 for validation), and again, 7/8 cases of the validation sub-cohort were correctly classified. QRT-PCR was then utilized to classify further 12 cases (7 cases defined as PCA2) not employed for GEP analysis. Overall, in the 42 cases, 23 cases were considered as PCA2 with the GEP/QRT-PCR approach. iv) Clinical/biological correlations. No association was found between the 6-gene signature and IGHV status (22/30 unmutated IGHV cases) or between the signature and the overexpression of SOX11 (17/30 cases over the median value). In addition, no association was found with the presence of the main recurrent mutations of the ATM, BIRC3, CCND1, KMTD2, NOTCH1, TP53, TRAF2, WHSC1 genes. Finally, an "ad-interim" analysis of progression free survivals (PFS) (Cortelazzo et al EHA, 2015) suggested a trend for a shorter PFS (2-years PFS 45% vs 72%, p=0.08) for cases classified as PCA2 by the GEP/QRT-PCR approach. v) 6-gene signature and sensitivity to the BCR inhibitor ibrutinib. The finding that PCA2 cases overexpressed BCR-related genes and had a more aggressive clinical course prompted us to investigate the 6-gene signature in the context of ibrutinib sensitive/resistant MCL cell lines. To do this, the proliferation rate of the MCL cell lines REC1, JEKO1, UPN1, GRANTA, JVM2, Z138 was investigated either in presence or in absence of ibrutinib 10 nanoM for 7 days. REC1, JEKO1 were selected as responsive by showing ≥80% inhibition upon ibrutinib. Of note, responsive cell lines showed higher expression levels of the 6-gene signature then the resistant counterpart, as evaluated by QRT-PCR. Conclusions. A novel 6-gene expression signature related to the BCR pathway has been found to characterize MCL cells with peculiar clinical/biological features and sensitivity to BCR inhibitors. Disclosures Luminari: Roche: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Teva: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2015
26. The Prognostic Role of Cell of Origin Profile and Myc Expression Assessed By Immunohistochemistry in Young High-Risk Patients with Diffuse Large B-Cell Lymphoma (DLBCL): Results of First-Line Randomized BIO-DLCL04 Trial of Fondazione Italiana Linfomi (FIL)
- Author
-
Caterina Stelitano, Manuel Gotti, Marco Ladetto, Simona Righi, Claudio Agostinelli, Domenico Novero, Andrea Evangelista, Gianluca Gaidano, Maurizio Martelli, Giuseppe Rossi, Stefano Pileri, Monica Balzarotti, Umberto Vitolo, Angelo Michele Carella, Emanuele Angelucci, and Annalisa Chiappella
- Subjects
medicine.medical_specialty ,Pathology ,business.industry ,Immunology ,Hazard ratio ,Histology ,Cell Biology ,Hematology ,BCL6 ,medicine.disease ,Biochemistry ,Gastroenterology ,Lymphoma ,Internal medicine ,medicine ,Immunohistochemistry ,Rituximab ,Risk factor ,business ,Diffuse large B-cell lymphoma ,medicine.drug - Abstract
Background. The prognostic role of cell of origin profile (COO) assessed by immunohistochemistry (IHC) is controversial in Rituximab era. FIL conducted a phase III randomized trial aimed at investigating the benefit of intensification with high dose therapy plus autotransplant compared to R-dose-dense therapy as first line in young DLBCL at poor risk (aa-IPI 2-3). Clinical results were reported (Vitolo, ASH 2012). The aim of BIO-DLCL04 was to correlate the biological markers with PFS. Patients and Methods. From 2005 to 2010, 412 untreated DLBCL at aa-IPI 2-3 were enrolled. Central histology revision was mandatory and 13 patients were excluded due to different histologies. Biological markers were analyzed on DLBCL NAS; COO analysis was performed by IHC and cases were classified in germinal center (GC) and non-GC according to Hans' algorithm; COO determined by gene expression profile using the NanoString® nCounter® Analysis System based on 20-gene assay (Lymph2Cx) using formalin fixed paraffin embedded tissue is ongoing; BCL2, BCL6 and MYC anomalies were tested by IHC; final analysis by fluorescent in situ hybridization (FISH) is ongoing. Cases were deemed positive if at least 30% of lymphoma cells were stained with each antibody (with the exception of at least 40% for MYC). Results. At the time of this analysis, 223 DLBCL NAS were analyzed: 131 non-GC and 92 GC; BCL2, BCL6 and MYC anomalies were tested in 196, 74 and 107 cases respectively. Clinical characteristics for non-GC vs GC were: median age 51 years for both, male 49% vs 45%, aa-IPI 3 15% vs 25%, bone marrow involvement (BM) 16% vs 24%. R-HDC was performed in 45% of non-GC patients and in 49% of GC. Complete response was recorded in 105 (80%) non-GC patients and in 62 (67%) GC. At a median follow-up of 49 months, the 3-year PFS for non-GC vs GC was 75% (95% CI: 67-82) vs 57% (95% CI: 46-67) with crude hazard ratio, HR 0.55 (0.35-0.87), p.01 and adjusted (for age, gender, aa-IPI, BM) aHR 0.56 (0.35-0.88), p.013. No significant differences by treatment were reported. Overexpression of MYC by IHC had a relevant prognostic impact, with aHR 1.84 (0.99-3.44), p.054. By IHC, 3-years PFS for double negative vs single BCL2 or MYC overexpression vs double positive, was 85% vs 68% vs 51% respectively, with an aHR for double expressors compared to double negative of 3.91 (1.13-13.53), p.031. At the time of the present report, FISH analysis was conducted in 88 cases: 43 were triple negative, 37 single hit and 8 double/triple hit. By FISH, 3-years PFS for triple negative vs single hit vs double/triple hit was 74% vs 84% vs 25% respectively, with an aHR for double/triple hit compared to triple negative of 5.73 (2.05 to 16.02), p.001. Conclusions. In conclusion, with the limit of the analysis performed by IHC based on Hans' algorithm, BIO-DLCL04 showed an unexpected better outcome for non-GC compared to GC, irrespective of treatment arm. The ongoing analysis conducted by Nanostring will be more informative. The overexpression of MYC was an unfavourable risk factor, mainly if associated with BCL2 overexpression, irrespective of type of treatment. Moreover, double/triple hit patients represent a subgroup with extremely poor prognosis. High dose therapy plus autotransplant was not able to reverse the inferior outcome of neither double expressors nor double hit patients and new strategies are deemed for these poor prognosis patients. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
27. Library Preparation Is the Major Factor Affecting Differences in Results of Immunoglobulin Gene Rearrangements Detection on Two Major Next-Generation Sequencing Platforms
- Author
-
Jan Trka, Monika Brüggemann, Jeremy Hancock, Grazia Fazio, Simone Ferrero, Henrik Knecht, Simona Songia, Jack Bartram, Andrea Grioni, Anton W. Langerak, Dietrich Herrmann, John Moppett, Vojtech Bystry, Giovanni Cazzaniga, Cristina Jiminez, Ramón García-Sanz, Eva Fronkova, Michaela Kotrova, Christiane Pott, Marco Ladetto, Elisa Genuardi, and Nikos Darzentas
- Subjects
Genetics ,Serial dilution ,Immunology ,Cell Biology ,Hematology ,Gold standard (test) ,Ion semiconductor sequencing ,Biology ,Amplicon ,Biochemistry ,Minimal residual disease ,DNA sequencing ,Deep sequencing ,Primer (molecular biology) - Abstract
Minimal residual disease (MRD) assessment via next generation sequencing (NGS) of immunoglobulin (Ig) and T-cell receptor (TR) gene rearrangements for lymphoid malignancies is currently under extensive development. NGS MRD has a potential to overcome the limitations of current techniques; laboriousness and difficult interpretation of qPCR for Ig/TR and low sensitivity of flow cytometry. However, amplicon-based NGS MRD has potential pitfalls that have to be addressed before it can be safely introduced for clinical decision making. Multi-center concordance in the experimental setting, quality control and interpretation of the results need to be achieved in order to surpass the advantages of qPCR, which is currently rigorously standardized within the EuroMRD consortium. Our aim was to test the stability and reproducibility of an optimized Ig heavy chain (IGH) based NGS approach for MRD assessment in a multi-center setting within the EuroClonality NGS Consortium on two different sequencing platforms. A one-step PCR library preparation approach was tested in seven institutions (Kiel, Salamanca, Milano, Bristol, London, Prague, Torino). Serial dilutions (10-1 to 10-5) of diagnostic DNA into polyclonal DNA as well as follow-up samples of 30 B-cell precursor ALLs with known complete IGH rearrangements were sequenced on the MiSeq. Serial dilutions of five different diagnostic ALL samples and libraries from polyclonal control were sequenced in parallel on both the MiSeq and Ion Torrent platforms. All samples were spiked with pre-defined copy numbers of five reference IGH sequences as a calibrator. FR2 primers, harboring platform-specific sequencing adapters, were used during the one-step PCR with 500ng of DNA per sample (75,000 copies). Negative and positive controls (27 pooled B-cell lines) were used for testing assay stability and reproducibility among the labs. Purpose-built bioinformatics methods were applied to analyze data. MRD results were compared to results of EuroMRD-based qPCR results. A total of 333 libraries were sequenced in 29 deep sequencing runs producing 194 million reads. The IGH gene rearrangements of all 27 pooled positive B-cell line controls were identified in all centers. NGS MRD analysis in 116 ALL follow-up samples revealed MRD positivity in 69/116 samples vs. 66/116 samples in qPCR, with discrepancies concerning samples with low MRD (R2=0.81). The dilution experiments gave similar results for both platforms, with a minimum sensitivity of 10-4 (as currently required by most treatment protocols using qPCR) for all tested assays. The correlation between MRD levels obtained by the two NGS platforms was good (R2=0.84). Ratios of reads containing reference IGH sequences were highly consistent in intra- and inter-laboratory analyses, independent of the total number of reads in the sample. When comparing platforms, in 10-1 dilution samples sequenced on MiSeq the ratio of reads harboring reference sequences was 2.1 to 2.7 times lower than in remaining dilutions, while on the Ion Torrent it was only 0.9 to 1.3 times, reflecting the competition with the leukemic clone. The correlation of the amounts of spiked-in sequences with the representation of reads harboring these sequences was slightly better for the Ion Torrent (R2=0.88) than for the MiSeq (R2=0.79). Amplification efficiency of each primer was checked by analyzing libraries from healthy polyclonal control. All primer sequences were present in all samples on both platforms, however, the differences between four libraries prepared from the same sample sequenced on the MiSeq were 2.6 times higher than in one library from this sample sequenced in five replicates on the Ion Torrent. The newly developed IGH assay shows robust intra and inter-laboratory reproducibility, which is the first step towards the safe use of this new MRD technique in a multi-center setting. The distribution of reference sequences and sequences of primers confirmed that the main source of differences between platform strategies is the library preparation and not the platform itself. Using the same amount of DNA, the sensitivity of the method is similar to qPCR. The performance and costs of the assay are similar for both the MiSeq and Ion Torrent. MRD analysis via NGS has therefore a great potential to replace qPCR as the gold standard for MRD-guided therapy in ALL, provided that thorough standardization can be achieved. Support: NV15-30626A, GBP302/12/G101. Disclosures Langerak: Roche: Other: Lab services in the field of MRD diagnostics provided by Dept of Immunology, Erasmus MC (Rotterdam); DAKO: Patents & Royalties: Licensing of IP and Patent on Split-Signal FISH. Royalties for Dept. of Immunology, Erasmus MC, Rotterdam, NL; InVivoScribe: Patents & Royalties: Licensing of IP and Patent on BIOMED-2-based methods for PCR-based Clonality Diagnostics..
- Published
- 2015
28. Telomere disrupts, CLL progresses
- Author
-
Marco Ladetto
- Subjects
immune system diseases ,hemic and lymphatic diseases ,Chronic lymphocytic leukemia ,Immunology ,medicine ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Telomere - Abstract
In this issue of Blood , Lin and colleagues address in detail the nature and severity of telomere disruption in chronic lymphocytic leukemia (CLL).[1][1] Using sophisticated methods for telomere length determination, the authors show that critical telomere shortening occurs in a proportion of CLL
- Published
- 2010
29. The adulthood of MRD detection in MCL
- Author
-
Marco Ladetto
- Subjects
Oncology ,medicine.medical_specialty ,Pediatrics ,business.industry ,Immunology ,Follicular lymphoma ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Predictive value ,Article ,law.invention ,body regions ,Randomized controlled trial ,Outcome predictor ,law ,Acute lymphocytic leukemia ,Internal medicine ,hemic and lymphatic diseases ,Disease remission ,medicine ,Mantle cell lymphoma ,Quantitative Real-Time Polymerase Chain Reaction ,business - Abstract
The prognostic impact of minimal residual disease (MRD) was analyzed in 259 patients with mantle cell lymphoma (MCL) treated within two randomized trials of the European MCL Network (MCL Younger and MCL Elderly trial). After rituximab-based induction treatment 106/190 evaluable patients (56%) achieved a molecular remission (MR) based upon blood and/or bone marrow (BM) analysis. MR resulted in a significantly improved response duration (RD) (87% vs. 61% patients in remission at 2 years, p=0.0043) and emerged to be an independent prognostic factor for RD (HR 0.4, 95% CI 0.1–0.9, p=0.027). MR was highly predictive for prolonged RD independent of clinical response (CR, CRu, PR) (RD at 2 years: 100% in BM MRD-negative CR and 88% in BM MRD-negative CRu/PR, compared to 78% in BM MRD-positive CR and 53% in BM MRD-positive CRu/PR, p=0.0015). Sustained MR during the post-induction period was predictive for outcome in MCL Younger after ASCT (RD at 2 years 100% vs. 65%, p=0.0007) and during maintenance in MCL Elderly (RD at 2 years: 76% vs. 36%, p=0.015). ASCT in MCL Younger patients increased the proportion of patients in MR from 55% prior to high dose therapy to 72% thereafter. Sequential MRD monitoring is a powerful predictor for treatment outcome in MCL.
- Published
- 2010
30. Efficacy of Reduced-Intensity Allogeneic Stem Cell Transplant after Brentuximab Vedotin in Patients with Hodgkin Lymphoma Relapsed after Autologous Transplant
- Author
-
Gioacchino Catania, Flavia Salvi, Maria jose Fornaro, Massimo Pini, Nicola Mordini, Francesco Zallio, Marco Ladetto, Guido Parvis, Alessandro Busca, and Benedetto Bruno
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Immunology ,Population ,Salvage therapy ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Tacrolimus ,Surgery ,Transplantation ,Graft-versus-host disease ,Autologous stem-cell transplantation ,Internal medicine ,medicine ,Progression-free survival ,Brentuximab vedotin ,education ,business ,medicine.drug - Abstract
Purpose: The prognosis of patients with Hodgkin lymphoma (HL) who relapse following autologous stem cell transplantation (ASCT) is generally poor, because salvage chemo-radiotherapy is able to produce only short-lasting remissions. In a previous study on a large patients' population (Sarina B et al, Blood 2010) was clearly demonstrated that the availability of a donor could significantly improve survival outcomes after allogeneic transplant, in particular for the subset of patients who reach a status of pre-transplant complete remission; however the application of this procedure is actually limited by a difficulty to obtain an objective response before alloSCT. It was recently shown that Brentuximab vedotin, a new generation antibody-drug conjugate, is able to induce nearly 30% of complete remission in HL patients relapsing after autologous transplant; therefore this agent could effectively work in relapses after ASCT as a platform to allow a better disease control in order to improve the outcome of the allografting procedure. Aims: Aim of this study was to retrospectively investigate if brentuximab vedotin administered as a salvage therapy in HL patients relapsing after ASCT could improve the efficacy of a subsequent reduced-intensity allogeneic transplant. Methods: Between August 2011 and September 2013, 11 patients underwent to allo-SCT at four hematologic Divisions of Northern Italy for HL relapse after ASCT, after brentuximab vedotin administered as a bridge to the allografting procedure. Median age was 32 years (range 21-61) and median number of prior regimens was 5, including ASCT. Patients received a median of 6 cycles (range 4-7) of brentuximab vedotin administered every 3 weeks as a 30-minute outpatient IV infusion; median time between the last dose of brentuximab vedotin and the allo-SCT was 1 month (range 1-5 months). No patient experienced progression during treatment with brentuximab vedotin (4 complete remissions and 7 partial remission/stable disease). All but one patients did not have a HLA identical sibling, so they required a matched unrelated (6 patients), haploidentical donor (3 patients) or cord blood (1 patient); peripheral stem cells were the source in patients with HLA identical sibling or unrelated, whereas bone marrow was used in the haplo setting. Reduced-intensity was the conditioning regimen performed in all patients. Post transplant cyclophosphamide plus mycophenolate mofetil/tacrolimus was the graft-versus-host disease (GVHD) prophylaxis of the haplo setting, while Metotrexate/cyclosporine was administered in the other patients. Patients were monitored for engraftment and infectious complications per institutional standards; survival outcomes were defined according to the European Blood and Marrow Transplantation (EBMT) criteria for survival analysis. Results: One patient had primary graft failure with autologous reconstitution; she was a patient with a high body-mass index, in whom the drug's doses of the conditioning regimen were underestimated in order to avoid excessive toxicity. All the other patients engrafted; median time to neutrophil recovery was 20 days (range 15-26). At a median follow-up of 12 months, the 2-year progression free survival was 51%, 2-year Overall Survival was 61%. There were 2 toxic deaths, one for EBV reactivation and one for leuko-encefalopathy disease, resulting in a 2-year non-relapse mortality of 18%. Conclusions: These data suggest that brentuximab vedotin represents a promising salvage therapy in HL patients relapsed after ASCT. Despite the bias concerning the differences in source of stem cells and pre-transplant conditioning regimens, we showed that brentuximab could be administered in patients who are candidates to a RIC allo-SCT, with encouraging survival outcomes. Further clinical trials with larger number of patients and longer follow-up are recommended to confirm the promising role of brentuximab as a bridge to alloSCT. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
31. Prospective Molecular Monitoring of Minimal Residual Disease after Non-Myeloablative Allografting in Newly Diagnosed Multiple Myeloma
- Author
-
Luigia Monitillo, Simone Ferrero, Mario Boccadoro, Lucia Brunello, Paola Ghione, Silvia Cena, Luisa Giaccone, Roberto Passera, Benedetto Bruno, Daniela Barbero, Antonio Palumbo, Renato Fanin, Nicola Mordini, Francesca Patriarca, Federica Ferrando, Daniela Drandi, Paola Omedè, Moreno Festuccia, and Marco Ladetto
- Subjects
Melphalan ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Immunosuppression ,Cell Biology ,Hematology ,Gene rearrangement ,medicine.disease ,Biochemistry ,Gastroenterology ,Minimal residual disease ,Surgery ,Transplantation ,Median follow-up ,Internal medicine ,medicine ,Cumulative incidence ,business ,Multiple myeloma ,medicine.drug - Abstract
Background Evaluation of minimal residual disease (MRD) by molecular methods is used as a surrogate of potential eradication of myeloma cells. Prolonged high-rates of molecular remission were reported after myeloablative allografting. However, recent studies on consolidation therapy after an autograft reported 18% of PCR negativity by qualitative nested PCR and a molecular response of 63% by quantitative RQ-PCR (Ferrero et al. Leukemia 2014). The aim of our study was that of evaluating depth of response by molecular methods induced by graft-vs.-myeloma following tandem autologous/non-myeloablative allografting in newly diagnosed myeloma. Patients and Methods. Twenty-six patients enrolled in prospective clinical trials (ClinicalTrial.gov Identifier: NCT-00702247, and NCT01264315) with a diagnostic bone marrow (BM) specimen suitable for immunoglobulin heavy-chain gene rearrangement sequencing were evaluated for MRD by qualitative nested PCR and quantitative RQ-PCR. A patient specific marker was generated for 19/26 (73%). Sensitivity of nested PCR was 3 neoplastic rearrangements in 106 normal cells while each RQ-PCR showed a standard curve correlation coefficient of at least 0.95 with a slope of 3.0 to 3.9 with minimum sensitivity of 10-5 (according to Euro-MRD criteria). Transplant plan consisted of a standard autograft (melphalan 200 mg/m2) followed by 200 cGy TBI and an allograft. Post-transplant donor lymphocyte infusions or maintenance/consolidation with new drugs were not allowed until clinical relapse. BM samples were collected at diagnosis, after the autograft, at month 1, 3, 6 after the allograft and then every 6 months or as clinically indicated. MRD negativity by nested-PCR or by RQ-PCR was defined as al least two consecutive samples scored negative. Results. At a median follow-up of 11.9 years (6.1-15.2) from diagnosis and 11.0 years (5.0-14.2) from the allograft, median overall survival (OS) for the entire patient cohort was not reached and median event-free survival (EFS) was 4 years from the allograft. Overall transplant-related mortality was 11.5% at 1 year and 15.4% at 5 years. Relapse was 3.8% at 1 year, 30.8% at 3 years and 34.6% at 5 years. Cumulative incidence of acute II-IV GVHD was 26.9%. At follow up, only 2 patients were on immunosuppression for limited chronic GVHD. In the 19/26 (73%) patients with a molecular marker, the rate of nested-PCR negativity remained low at month 3 (3/19, 16%) after the allograft and gradually increased at month 6 and 12 up to 44% (8/18) and 47% (7/15) respectively. By RQ-PCR analysis, the overall tumor reduction of the whole treatment was 13.80 ln and progressively increased through the autograft, the allograft and the post-transplant period (p Conclusions. G raft-vs-myeloma after non-myeloablative allografting reported prolonged rates of qualitative (nested PCR) molecular remissions similar to those after myeloablative allografting and higher than the recently reported combination of autografting and consolidation with anti-myeloma agents. Quantitative RQ-PCR analysis showed a deeper tumor reduction after non-myeloablative allografting than consolidation with new drugs. In the light of our results, it may be ethical to evaluate the combination of graft-vs-myeloma with newer anti-myeloma agents on larger series of young high risk and early relapsed patients where life expectancy is poor also in the era of new drugs. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
32. The Prognostic Value of MYC, BCL2 and BCL6 Overexpression Evaluated By Immunohistochemistry (IHC) in De-Novo Diffuse Large B Cell Lymphoma (DLBCL) Treated with Rituximab-CHOP
- Author
-
Federico Vittone, Roberto Freilone, Giovannino Ciccone, Paola Riccomagno, Giovanni Cametti, Domenico Novero, Annalisa Chiappella, Andrea Evangelista, Barbara Botto, Paola Ghione, Mattia Novo, Marco Ladetto, Umberto Vitolo, Chiara Ciochetto, and Alessia Castellino
- Subjects
Oncology ,medicine.medical_specialty ,Univariate analysis ,Proliferation index ,business.industry ,Immunology ,Hazard ratio ,Cell Biology ,Hematology ,CHOP ,medicine.disease ,Biochemistry ,International Prognostic Index ,immune system diseases ,Median follow-up ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Progression-free survival ,business ,neoplasms ,Diffuse large B-cell lymphoma - Abstract
Introduction : MYC, BCL2 and BCL6 overexpression, assessed by IHC, with the latter conferring a better prognosis, have been reported to be a prognostic factor in DLBCL, but data are not consistent and sometimes contradictory. The aim of the present study was to assess the prognostic impact of overexpression of MYC, BCL2, and BCL6 in a retrospective cohort of de-novo DLBCL, selected for an high proliferation index (MIB1 ≥70%), treated consecutively with R-CHOP regimen. Methods: Patients with de-novo DLBCL diagnosed between January 2010 and December 2013 were included into the study. Inclusion criteria were: high proliferation index MIB1 ≥ 70% and a full course of R-CHOP regimen. Paraffin-embedded tumor samples were collected and investigated using immunohistochemistry (IHC) for MYC, BCL2 and BCL6. Fluorescence in situ hybridization (FISH) is ongoing. MYC/BCL2+ or MYC/BCL6+ double expression cases were identified if they had rearrangements of MYC and BCL2 or BCL6. MYC immunochemistry was done on TMA sections using the antibody clone Y69. BCL2 and BCL6 staining had been evaluated previously at diagnosis. Tumor cells were defined positive for MYC and BCL2 or BCL6 protein expression by immunostaining if >40%, >40% and >25% of cells showed positive expression, respectively. Progression free survival curves (PFS) were estimated using the Kaplan-Meier method and compared between groups using the log-rank test and Cox models. Results : One hundred and sixty seven patients are evaluable for clinical characteristics and 69/167 had paraffin embedded tumor samples available for immunohistochemistry at the time of present analysis. Clinical characteristics of the 69 cases were: median age 66 years (IQR 57;73), 45 (65%) male, 47 (68%) stage III-IV, 35 (54%) with elevated LDH levels and 46 (67%) at International Prognostic Index (IPI) high intermediate or high risk. Overexpression of MYC was detected in 28 cases (41%), 50 (72%) and 38 (55%) showed BCL2 and BCL6 overexpression respectively. Nineteen (28%) cases showed MYC/BCL2+ and 17 (25%) MYC/BCL6+ double expression. With a median follow up of 26 months, the median 2-years PFS was 59%. Overexpression of MYC and BCL2 proteins and low expression of BCL6 were associated with an inferior 2-years PFS in univariate analysis: MYC- vs MYC+ 64% vs 55%; BCL2- vs BCL2+ 71% vs 56%; BCL6+ vs BCL6- 61% vs 54%. In a Cox multivariate regression model adjusted for IPI and age, MYC overexpression, BCL2 positivity and BCL6 negativity showed prognostic relevance as significant independent indicators with different risk (Hazard ratio 2.53 for MYC+, 2.08 for BCL2+ and 1.62 for BCL6-). Established that the three variable contributed with different risk in the multivariate analysis, an IHC sum additive score of 0-5 was calculated proportionally to the coefficient estimated (coefficient [Log hazard ratio] 0.92 for MYC+, 0.73 for BCL2+ and 0.48 for BCL6-), assigning an individual risk of 2 points for MYC or BCL2 positivity and 1 point for BCL6 negativity. Two years-PFS was significantly different between all separate groups (Hazard ratio for unit increase 1.57 95% CI 1.11-2.22, p=0.01). After pooling scores 0-1 (with or without BCL6), 2 (presence of MYC or BCL2 only), and 3-4-5 (MYC+/BCL6-, BCL2+/BCL6-, MYC+/BCL2+, MYC+/BCL2+/BCL6-) 2-yrs PFS rates were different across the three groups: 100% vs 64% vs 50% (log rank p= 0.04) (figure 1). Conclusion: Our data showed, with the limits of a small sample size, that MYC overexpression alone or with high expression of BCL2 and/or low expression of BCL6 correlates with a worse prognosis independently by IPI score in a cohort of DLBCL selected for high proliferation index and treated with R-CHOP. Assessment of MYC, BCL2 and BCL6 expression by IHC represents a rapid, inexpensive, and reproducible technique. These results need to be confirmed in our complete series of 167 patients (analysis ongoing) and validated prospectively in a larger cohort, using standardized staining and scoring methodologies. Thus, MYC and BCL2 represent relevant biomarkers that should be tested in future clinical trials using novel effective and targeted agents in order to improve the prognosis of DLBCL. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
33. Prognostic Impact of p190 and p210 Co-Expression at Diagnosis in Chronic Myeloid Leukemia (CML) Patients Treated with Imatinib
- Author
-
Michela Salvio, Anna Baraldi, Marco Ladetto, Nicol Trincheri, Daniela Pietrasanta, Francesco Zallio, Gioacchino Catania, Federico Monaco, Massimo Pini, Salvi Flavia, Maria Teresa Corsetti, and Depaoli Lorella
- Subjects
Pathology ,medicine.medical_specialty ,Myeloid ,business.industry ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Gastroenterology ,Dasatinib ,Leukemia ,medicine.anatomical_structure ,Imatinib mesylate ,Nilotinib ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,Internal medicine ,medicine ,business ,Sokal Score ,medicine.drug - Abstract
Background: Expression of p190 BCR-ABL mRNA is generally considered to be confined to patients with acute lymphoid or more rarely myeloid leukemias, whereas p210 BCR-ABL mRNA is the hallmark of CML. In reality it is not uncommon the presence of p190 m-RNA in p210 CML in chronic phase, due to alternative or missplicing1. Its presence seems to have no impact on prognosis in the pre-TKI era, although it may be expression of genomic instability. Aim: Primary object of this study was to investigate if the co-expression might influence the rate of early outcome surrogate endpoints such as such as early complete cytogenetic response in patients treated with imatinib. The secondary endpoint was the evaluation of failure free survival (FFS) measured from the start of imatinib to the date of any of the following events: progression to accelerated or blastic phase, death for any cause and switch to nilotinib/dasatinib for resistance or intolerance. Methods: Were evaluated patients with CML in chronic phase treated with imatinib at our institution. We excluded cases with less than one year of treatment and/or treated with other TKIs or conventional chemotherapy.The fusion transcripts BCR-ABL were evaluated at diagnosis in peripheral blood by NESTED-PCR2 and the cytogenetic response were evaluated in bone marrow cells with G-banding technique and fluorescent in situ hybridization (FISH)3. The patients were divided into two groups, "double transcripts" (DT) and "single transcript" (ST). All patients received imatinib 400 mg/die. Results: A total of 56 patients were analyzed. The median age of patients was 58 years (range 28-80 years) and 35 (62%) were male. Twenty patients (36%) were DT and thirty-six (64 %) ST. The distribution according to Sokal score was: 7 (35%), 8 (40%) and 5 (25%) patients for low, intermediate and high risk in the DT, whereas 18 (50%), 15 (42%) and 3 (8%) low, intermediate and high risk in ST, respectively. The complete cytogenetic response at 3 months was achieved in 2 patients with DT and 7 patients with ST (10% vs 19% p 0.35), at 6-month complete cytogenetic response was achieved in 8 patients with DT and 27 patients with ST (40% vs 75% p 0.01) (Table 1). After median follow-up of 1966 days, the FFS was significantly different between the DT and ST (55% vs 5 % p< 0.001) (figure 1), 11 patients in the DT group and 5 patients in ST group had shift to TKI 2¡ generation (55% vs 14% p 0.001) and 4 patients in DT group not achieved complete cytogenetic response. Summary/Conclusion: In our study the co-expression of p190 and p210 BCR-ABL transcripts influences the early cytogenetic response to imatinib and suggesting the need for a larger validation study Reference 1). van Rhee F, Hochhaus A, Lin F, Melo JV, Goldman JM, Cross NC. : "p190 BCR-ABL mRNA is expressed at low levels in p210-positive chronic myeloid and acute lymphoblastic leukemias."Blood. 1996 15;87:5213-7. 2). Hermans A, Selleri L, Gow J, Wiedemann L, Grosveld G: "Molecular analysis of the Philadelphia translocation in chronic myelocytic and acute lymphocytic leukemia." Leukemia 2:628,1988. 3) Landstrom AP, Tefferi A.: "Fluorescent in situ hybridization in the diagnosis, prognosis, and treatment monitoring of chronic myeloid leukaemia".Leuk Lymphoma.2006;47:397-402. Figure 1: Failure Free Survival: Figure 1:. Failure Free Survival: Table 1 : Complete Cytogenetic response: Figure 2 Figure 2. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
34. Telomere length correlates with histopathogenesis according to the germinal center in mature B-cell lymphoproliferative disorders
- Author
-
Daniela Drandi, Roberto Francese, Mara Compagno, Irene Ricca, Marco Ladetto, Loredana Santo, Barbara Mantoan, Paola Francia di Celle, Federica De Marco, Maria Dell’Aquila, Mario Boccadoro, Corrado Tarella, Monica Astolfi, Gloria Pagliano, Sonia Vallet, Marco Pagano, Alberto Rocci, A Cuttica, and Carlo Marinone
- Subjects
Telomerase ,B-Lymphocytes ,Leukemia ,Chronic lymphocytic leukemia ,Immunology ,Restriction Mapping ,Follicular lymphoma ,Germinal center ,Cell Biology ,Hematology ,Biology ,Telomere ,medicine.disease ,Biochemistry ,Molecular biology ,Lymphoproliferative Disorders ,Lymphoma ,medicine ,Humans ,Mantle cell lymphoma ,Multiple Myeloma ,Diffuse large B-cell lymphoma ,Burkitt's lymphoma - Abstract
In this study we investigated telomere restriction fragment (TRF) length in a panel of mature B-cell lymphoproliferative disorders (MBCLDs) and correlated this parameter with histology and histopathogenesis in relation to the germinal center (GC). We assessed 123 MBCLD samples containing 80% or more tumor cells. TRF length was evaluated by Southern blot analysis using a chemiluminescence-based assay. GC status was assessed through screening for stable and ongoing somatic mutations within the immunoglobulin heavy-chain genes. Median TRF length was 6170 bp (range, 1896-11 200 bp) and did not correlate with patient age or sex. TRF length was greater in diffuse large cell lymphoma, Burkitt lymphoma, and follicular lymphoma (medians: 7789 bp, 9471 bp, and 7383 bp, respectively) than in mantle cell lymphoma and chronic lymphocytic leukemia (medians: 3582 bp and 4346 bp, respectively). GC-derived MBCLDs had the longest telomeres, whereas those arising from GC-inexperienced cells had the shortest (P < 10-9). We conclude that (1) TRF length in MBCLD is highly heterogeneous; (2) GC-derived tumors have long telomeres, suggesting that minimal telomere erosion occurs during GC-derived lymphomagenesis; and (3) the short TRF lengths of GC-inexperienced MBCLDs indicates that these neoplasms are good candidates for treatment with telomerase inhibitors, a class of molecules currently the subject of extensive preclinical evaluation. (Blood. 2004;103:4644-4649)
- Published
- 2004
35. Reduced-intensity conditioning followed by allografting of hematopoietic cells can produce clinical and molecular remissions in patients with poor-risk hematologic malignancies
- Author
-
Moreno Tresoldi, Alessandro Pileri, Claudio Bordignon, Michele Falda, Corrado Tarella, Maria R. Sajeva, Francesco Zallio, Anna Dodero, Marco Ladetto, Moira Lucesole, Marco Bregni, Fabio Ciceri, Claudia Voena, Fabio Benedetti, Attilio Olivieri, Alessandro Rambaldi, Paolo Corradini, Alessandro M. Gianni, Corradini, P, Tarella, C, Olivieri, A, Gianni, Am, Voena, C, Zallio, F, Ladetto, M, Falda, M, Lucesole, M, Dodero, A, Ciceri, Fabio, Benedetti, F, Rambaldi, A, Sajeva, Mr, Tresoldi, M, Pileri, A, Bordignon, Claudio, and Bregni, M.
- Subjects
Adult ,Male ,medicine.medical_specialty ,Neoplasm, Residual ,Transplantation Conditioning ,Immunology ,Graft vs Host Disease ,ThioTEPA ,Lymphoma, Mantle-Cell ,Biochemistry ,Gastroenterology ,Polymerase Chain Reaction ,Recurrence ,Internal medicine ,Medicine ,Autologous transplantation ,Humans ,Transplantation, Homologous ,Cyclophosphamide ,Aged ,Transplantation Chimera ,Anemia, Refractory, with Excess of Blasts ,business.industry ,Graft vs Tumor Effect ,Remission Induction ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Donor Lymphocytes ,Fludarabine ,Surgery ,Transplantation ,Survival Rate ,Regimen ,Methotrexate ,Hematologic Neoplasms ,Lymphocyte Transfusion ,Cyclosporine ,Mantle cell lymphoma ,Female ,business ,Refractory anemia with excess of blasts ,Thiotepa ,Vidarabine ,medicine.drug - Abstract
A reduced-intensity conditioning regimen was investigated in 45 patients with hematologic malignancies who were considered poor candidates for conventional myeloablative regimens. Median patient age was 49 years. Twenty-six patients previously failed autologous transplantation, and 18 patients had a refractory disease at the time of transplantation. In order to decrease nonrelapse mortality, and enhance the graft-versus-tumor effect, a program was designed in which a reduced conditioning with thiotepa, fludarabine, and cyclophosphamide was associated with programmed reinfusions of donor lymphocytes for patients without graft-versus-host disease (GVHD), not achieving clinical and molecular remission after transplantation. GVHD prophylaxis consisted of cyclosporine A and methotrexate. Seventeen patients received marrow cells and 28 received mobilized hematopoietic cells. All patients engrafted. The probability of grades II-IV and III-IV acute GVHD were 47% and 13%, respectively. The probability of nonrelapse mortality, progression-free survival, and overall survival were 13%, 57%, and 53%, respectively. Thirteen patients in complete remission had a polymerase chain reaction marker for minimal disease monitoring; 10 achieved molecular remission after transplantation. Nine patients received donor lymphocytes: one patient with mantle cell lymphoma had a minimal response, one patient with refractory anemia with excess of blasts in transformation achieved complete remission, and 7 patients did not respond. At a median follow-up of 385 days (range, 24 to 820 days), 25 patients (55%) were alive in complete remission. Although longer follow-up is needed to evaluate the long-term outcome, the study shows that this regimen is associated with a durable engraftment, has a low nonrelapse mortality rate, and can induce clinical and molecular remissions.
- Published
- 2002
36. Inhibition Of Histone Deacetylases With Panobinostat As a Treatment For Relapsed Or Refractory Diffuse Large B-Cell Lymphoma: A Phase II Study By The Fondazione Italiana Linfomi
- Author
-
Francesco Zaja, Annalisa Chiappella, Gianluca Gaidano, Pier Paolo Piccaluga, Stefano Volpetti, Stefano Pileri, Manuela Zanni, Flavia Salvi, Alessandra Tucci, Alfonso Maria D'Arco, Alessandro Levis, Pier Luigi Zinzani, Angelo Michele Carella, Silvia Franceschetti, Marco Ladetto, Gian Matteo Pica, Renato Fanin, and Andrea Evangelista
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Phases of clinical research ,Salvage therapy ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Surgery ,chemistry.chemical_compound ,chemistry ,Internal medicine ,Multicenter trial ,Panobinostat ,medicine ,Progression-free survival ,business ,Diffuse large B-cell lymphoma - Abstract
Backgrounds Histone deacetylases (DACs) are involved in chromatin structure regulation and function. Treatment with DACs inhibitors leads to the activation or repression of genes regulating apoptosis, proliferation, differentiation, angiogenesis, immune responses. These agents resulted to be active for the treatment of T and B-cell lymphoma and other haematological malignancies. Previous in vitro studies underlined the possible pathophysiological role of DACs in diffuse large B-cell lymphoma (DLBCL). In FIL-PanAL10 we evaluate the therapeutic activity and safety of Panobinostat, a potent pan-DACs inhibitor, in patients with relapsed or refractory (R/R) DLBCL. Methods FIL-PanAL10 (NCT01523834) is a phase II, prospective multicenter trial of the Fondazione Italiana Linfomi (FIL). Enrolled patients are ≥ 18 years old with R/R DLBCL, following ≥ 1 line of chemo-immunotherapy (R-CHOP) including high dose therapy with autologous stem cell support (ASCT) in eligible patients. Patients with > 5 prior systemic lines of treatment, CNS involvement, HIV positivity, impaired cardiac function (according to the protocol) are excluded. Patients are scheduled to receive Panobinostat po 40 mg three-times every week as part of a 4-weeks treatment cycle up to disease progression, unacceptable toxicity, or patient’s refusal. The primary endpoint of the study is to explore the antitumor activity of Panobinostat in terms of overall response (ORR) according to the Cheson criteria 1999. Secondary objectives are the evaluation of complete response (CR), time to response (TTR), progression free survival (PFS), overall survival (OS) and safety. Exploratory objectives evaluate the predictive role of pharmacogenetics, immunohistochemical and specific gene expression in relation to the response to Panobinostat; for this aim a new lymph node or other pathologic tissue biopsy is requested before starting treatment. With the null (P0) and P1 hypothesis of overall response (ORR) corresponding to Results Between February 2011 and May 2013, 23 patients were enrolled on this study. Patients’ median age is 73 years (range 44-83 years); 7 (30%) patients received ≥ 3 previous lines of chemotherapy. Five patients responded to Panobinostat (ORR= 22%) including 3 CR (13%) and 2 (9%) partial response (PR): 1 patient had (4%) stable disease (SD). TTR was 2.5 months (range 2-3 months ). All 6 patients with ORR or SD are still in treatment with Panobinostat after 4, 5, 12, 12, 15, 24, months; 17 patients discontinued therapy because of progression (15) or side effects (2). After a median period of observation of 11 months from the beginning of treatment, 1-year PFS and OS are 22% and 28%, respectively. Most common observed grade 3-4 adverse events were hematological and included thrombocytopenia (88%) and neutropenia (34%); grade 3-4 diarrhoea was present in 3 (13%). The therapeutic schedule was modified from the three times week to three times every other week in 15 patients; a further dose reduction from 40 mg to 30 mg resulted to be necessary in 8 patients. The analysis of exploratory biologic objectives and of their relationship with outcome is still under investigation and will be ready for December 2013. Conclusions The preliminary results of this study indicates that Panobinostat is an active salvage therapy in nearly 20% of heavily pretreated R/R DLBCL patients; the relatively short TTR observed allows a prompt shift to other rescue treatments in non responders. The analysis of biologic biomarkers will hopefully better address Panobinostat therapy to a specific biologic subgroup. Disclosures: Zaja: Mundipharma: Consultancy, Honoraria; Roche: Consultancy, Honoraria; Celgene: Consultancy; GSK: Consultancy, Honoraria; Amgen: Consultancy. Off Label Use: Panobinostat in DLBCL.
- Published
- 2013
37. A Comparative Analysis Of Next-Generation Sequencing and Real-Time Quantitative PCR For Minimal Residual Disease Detection In Follicular Lymphomas
- Author
-
Barbara Mantoan, Elisa Genuardi, Marco Ladetto, Monika Brüggemann, Luigia Monitillo, Christiane Pott, Heiko Trautmann, Michael Kneba, and Malek Faham
- Subjects
Genetics ,Massive parallel sequencing ,Immunology ,Breakpoint ,Consensus PCR ,Follicular lymphoma ,Somatic hypermutation ,Context (language use) ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Minimal residual disease ,Germline mutation ,hemic and lymphatic diseases ,medicine - Abstract
Background Detection of minimal residual disease (MRD) by t(14;18) Real-Time Quantitative (RQ) PCR is an important tool during clinical follow-up in patients (pts) with follicular lymphoma (FL). However, only the major breakpoint region (MBR) and minor cluster region (mcr) breakpoints have been currently exploited as MRD targets. Therefore, only 50% to 65% of pts can be assessed by t(14;18) RQ-PCR. Alternative targets such as the immunoglobulin heavy chain variable region (IGH) can be used only with limitations due to somatic hypermutation. IGH-based next-generation sequencing (NGS) might provide an alternative approach and provide increased sensitivity, specificity, accuracy and reproducibility. We performed a comparison of the two approaches in 29 FL pts. Patients and Methods Overall, 206 samples (85 bone marrow, 114 peripheral blood, 6 stem cell aliquots and one lymph node sample) were investigated from 29 FL pts in which RQ-PCR based MRD analysis had been performed in the context of prospective clinical trials. Overall, 33 dx and 173 follow-up (FU) samples were analyzed. 23/29 pts had a PCR detectable t(14;18) rearrangement, 5 pts had a clonal IGH rearrangement only and one patient had no marker by consensus PCR. NGS was performed at Sequenta in South San Francisco. Using universal primer sets, we amplified IGH variable, diversity, and joining gene segments from genomic DNA. Amplified products were sequenced to obtain a high degree of coverage and analyzed using standardized algorithms for clonotype determination. Tumor-specific clonotypes were identified for each patient based on their high-frequency in the dx sample and then quantitated in FU samples. Overall comparability of MRD results by RQ-PCR and NGS was assessed using correlation analysis. A positive/negative discordance between two results was defined as major when the positive result was >1E-05 and minor when ≤1E-05; a quantitative discordance was defined as the presence of two positive results with a quantitative discrepancy >1 log. Results All 29 pts were evaluable with at least one method. IGH NGS identified an index clone in 22/29 cases. Seven patients pts were only quantifiable by t(14;18) RQ-PCR while IGH NGS did not identify an index clone for sequencing. In all but one dx sample demonstrated low level lymphoma infiltration with MRD level below 10-3. In one of these cases, IG kappa could be successfully sequenced for MRD indicating that not only low level MRD but also somatic mutation of IGH is a potential pitfall for MRD detection by NGS in FL. 15 pts were evaluable for MRD by t(14;18) RQ-PCR and NGS. In 97 FU samples, a significant concordance between MRD methods could be demonstrated (r2=0.80) (p In 5 t(14;18) negative cases and one with an unusual large t(14;18) rearrangement MRD was quantified by IGH-RQ-PCR using cloned plasmids and IGH NGS. In all cases both methods also showed excellent concordance. One advantage of the IGH NGS approach is its ability to detect different IGH sequences generated by the somatic hypermutation process. We have seen in some patients a dynamic picture of the IGH clonotypes with the rise and fall of different clonotypes at different time points (Fig 1). Conclusions NGS represents a feasible tool for IGH-based MRD monitoring that allows analysis of a larger group of FL pts. Our results show that the two methods have a high level of correlation. Lymphoma infiltration of dx samples and somatic mutation of IGH is a critical point for identification of the tumor-specific clonotypes by NGS, therefore different MRD methods should complement each other to allow MRD assessment for the majority of pts. Furthermore IGH NGS sequencing has the potential to detect and track IGH evolution in FL. Disclosures: Kneba: Roche: Consultancy, Research Funding. Faham:Sequenta, Inc.: Employment, Patents & Royalties.
- Published
- 2013
38. Final Results Of Phase II Study Of Lenalidomide Plus Rituximab-CHOP21 In Elderly Untreated Diffuse Large B-Cell Lymphoma Focusing On The Analysis Of Cell Of Origin: REAL07 Trial Of The Fondazione Italiana Linfomi
- Author
-
Marcello Gaudiano, Marco Ladetto, Ileana Baldi, Gianluca Gaidano, Angela Congiu, Martin Dreyling, Barbara Botto, Giorgio Inghirami, Pier Paolo Fattori, Giovannino Ciccone, Anna Marina Liberati, Annalisa Chiappella, Giuseppe Rossi, Alessandra Tucci, Alfonso Zaccaria, Anna Lia Molinari, Manuela Zanni, Pier Luigi Zinzani, Angelo Michele Carella, Silvia Franceschetti, Michele Spina, Alessia Castellino, Flavia Salvi, Vincenzo Pavone, and Umberto Vitolo
- Subjects
medicine.medical_specialty ,business.industry ,Standard treatment ,Immunology ,Phases of clinical research ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Gastroenterology ,Surgery ,law.invention ,International Prognostic Index ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Rituximab ,Stage (cooking) ,business ,Diffuse large B-cell lymphoma ,Lenalidomide ,medicine.drug - Abstract
Introduction The standard treatment for elderly untreated diffuse large B-cell lymphoma (DLBCL) is RCHOP21, however up to 40% of patients experienced failures. Lenalidomide showed activity in heavily pretreated DLBCL and in vivo and in vitro data demonstrated a synergism with rituximab. In the phase I trial REAL07 (Chiappella et al, Haematol 2013), FIL demonstrated that the association of LRCHOP21 was feasible in elderly untreated DLBCL and identified 15 mg lenalidomide from day 1 to day 14 as the maximum tolerated dose in combination with RCHOP21. Patients and methods. The phase II trial REAL07 was designed based on Simon's two stage design to demonstrate an improvement of overall response rate (ORR) of 15% in LRCHOP21 compared to 70% of standard RCHOP21. Secondary endpoints were progression-free survival (PFS), overall survival (OS), event-free survival (EFS) and to correlate outcome with cell of origin (COO) profile. Response was evaluated according to 2007 Cheson criteria. Inclusion criteria were: age 60-80 FIT at the comprehensive geriatric assessment; untreated CD20+ DLBCL; Ann Arbor stage II/III/IV; international prognostic index (IPI) at low-intermediate/intermediate-high/high (LI/IH/H) risk. Treatment plan was: RCHOP21 plus 15 mg lenalidomide from day 1 to 14 for 6 courses. All cases were centrally reviewed by expert pathologist; COO profile analysis was conducted with immunohistochemistry according to Hans' algorithm and with gene expression profile (DASL assay). Results. From April 2010 to May 2011, 49 patients were enrolled. Clinical characteristics were: median age 69 years (range 61-80); stage III/IV 43 (88%), IPI IH/H 30 (61%). At the end of 6 LRCHOP21, ORR was 92%. Complete remissions (CR) were 42 (86%) and partial remission 3 (6%); 3 patients (6%) did not respond and one (2%) died for homicide. At a median follow-up of 28 months, 2-year OS was 92% (95% CI: 79-97), 2-year PFS was 80% (95% CI: 64-89) and 2-years EFS was 70% (95% CI: 55-81); 2-year PFS for IPI LI was 89% (95% CI: 62-97) and for IPI IH 76% (95% CI: 47-90) and for IPI H 72% (95% CI: 36-90). Hematological and extra-hematological toxicities were mild, with no grade IV extra-hematological events and no toxic deaths during treatment. Of the 294 planned courses of LRCHOP21, 277 (94%) were administered; median dose of lenalidomide delivered was 1185 mg (94% of the planned dose); at least 90% of the planned dose of each drug was administered in 91% of the RCHOP21 courses. Median interval time between RCHOP21 courses was 21 days (range 19-48). All 49 cases underwent central pathology review and diagnosis of DLBCL was confirmed. Regarding COO analysis, tissue block or stained slides were collected in 40/49 (82%), of which 32 were adequate for analysis. At the time of this abstract, COO analysis was reported according to immunohistochemistry data; DASL analysis is ongoing. Clinical characteristics between germinal center (GCB, 16 patients) and non-GCB (16 patients) were superimposable, excepted for a majority of H IPI risk in non-GCB group (p 0.067). ORR for GCB and non-GCB were 88% (CR 81%) and 88% (CR 88%), respectively. At a median follow-up of 28 months, 2-year PFS was 71% (95% CI: 40-88) in GCB-group and 2-years PFS was 81% (95% CI: 51-93) in non-GCB-group (Figure 1). Conclusions. In conclusion, LRCHOP21 is effective, also in poor risk patients, namely in non-GCB subgroup. These encouraging data warrant a future phase III randomized trial comparing LRCHOP21 vs. RCHOP21 in untreated non-GCB DLBCL. Disclosures: Off Label Use: lenalidomide in first line DLBCL is off lable. drug provided free by Celgene. Vitolo:Roche: Speakers Bureau; Celgene: Speakers Bureau; Takeda: Speakers Bureau.
- Published
- 2013
39. Minimal Residual Disease Monitoring During Maintenance In Multiple Myeloma Patients
- Author
-
Alberto Rocci, Manuela Gambella, Paola Omedè, Daniela Drandi, Francesca Gay, Francesca Patriarca, Federica Cavallo, Maria Teresa Petrucci, Caravita Tommaso, Giulia Benevolo, Norbert Pescosta, Stefania Oliva, Massimo Offidani, Vittorio Montefusco, Anna Marina Liberati, Antonietta Pia Falcone, Stelvio Ballanti, Tonino Spadano, Tommasina Guglielmelli, Pellegrino Musto, Renato Zambello, Francesco Di Raimondo, Marco Ladetto, Mario Boccadoro, and Antonio Palumbo
- Subjects
Melphalan ,medicine.medical_specialty ,Cyclophosphamide ,business.industry ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Minimal residual disease ,Surgery ,Transplantation ,Autologous stem-cell transplantation ,Maintenance therapy ,Internal medicine ,Medicine ,business ,Multiple myeloma ,medicine.drug ,Lenalidomide - Abstract
Background The quality of response and the residual disease after treatment are important prognostic factors in several hematological diseases including multiple myeloma (MM). Several papers demonstrated that the deeper the response after treatment, the longer the survival. However few data are available on the monitoring of minimal residual disease (MRD) during the maintenance therapy in transplant eligible MM patients. Aims to evaluate the role of maintenance therapy in reducing MRD and the role of monitoring the response to predict clinical relapse. Patients and Methods newly diagnosed MM patients enrolled in the RV-MM-EMN-441 trial (NCT01091831) and achieving at least a very good partial response (VGPR) after consolidation were included in the study. Patients received 4 Lenalidomide-Dexamethasone (RD) courses as induction, Cyclophosphamide to mobilize bone marrow stem cells (BMSC) and then were randomized to receive 6 cycles of Cyclophosphamide-Lenalidomide-Dexamethasone (CRD) or Autologous Stem Cell Transplantation (ASCT) with Melphalan 200 mg/m2. All patients received maintenance therapy with Lenalidomide (R) or Lenalidomide-Dexamethasone (RD) until relapse. MRD analysis was performed in a single laboratory (University of Turin, Italy) using flow cytometry according to European Myeloma Network guideline (Rawstron AC, Haematologica 2008). Samples of bone marrow (BM) were collected at diagnosis, after consolidation, after 3 and 6 courses of maintenance and then every 6 months until clinical relapse. The samples were considered MRD +ve if ≥ 0.01% of PC were detected. Immunophenotypic (IF) relapse was defined as an increase of ≥ 25% in the amount of malignant plasma cells in BM compared to the previous determination. Results Fifty patients (27 female/23 male) with a median age of 57 yrs (40-65) entered the study. According to ISS, 27 patients were stage I, 15 stage II and 8 stage III. Fish risk profile was standard in 31 patients, high in 11 and not available in 8. Twenty-five patients received CRD as consolidation and 25 underwent ASCT. The median follow-up was 28.6 months. After consolidation 16 (32%) patients achieve a complete response (CR) and 34 (68%) a VGPR. MRD was negative in 19/48 (40%) patients, of which 12 received ASCT (out of 23, 52%) and 7 received CRD (out of 25, 28%). Patients receiving ASCT showed a lower value of residual cells (median 0.08%, range 0 – 1.00) compared to patients receiving CRD (median 0.5%, range 0 – 2.9%, p=0.0134). The lower MRD value was achieved after consolidation in 31 patients (62%), after 3 courses of maintenance in 6 patients (12%) and after 6 or more courses of maintenance in 13 patients (26%). The increase in quality of response was observed primarily in patients receiving CRD: the average amount of residual plasma cells in bone marrow was 71/uL after induction, lowering to 51/uL after 6 and 12 courses of maintenance therapy. Nine patients clinically relapsed after an average time of 25.6 months from the beginning of the therapy and in all patients this was anticipated by immunophenotypic relapse. Conclusion 1) consolidation therapy with ASCT determines a deeper response compared to CRD; 2) maintenance therapy can improve the quality of response, in particular in patients not receiving ASCT; 3) Immunophenotypic relapse anticipate the clinical relapse. These results suggest the possible role of MRD monitoring to better assess the response to therapy also during maintenance and as marker of early relapse. Disclosures: Ladetto: Celgene: Research Funding, Speakers Bureau; Janssen Cilag: Research Funding, Speakers Bureau; Mundipharma: Research Funding, Speakers Bureau; Roche: Research Funding, Speakers Bureau; Amgen: Research Funding, Speakers Bureau. Boccadoro:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen Cilag: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Palumbo:Amgen: Consultancy, Honoraria; Bristol-Myers Squibb: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Millenium: Consultancy, Honoraria; Onyx: Consultancy, Honoraria.
- Published
- 2013
40. Radioimmunotherapy In Relapsed/Refractory Mantle Cell Lymphoma Patients: Final Results Of a European MCL Network Phase II Trial
- Author
-
Wolfgang Hiddemann, Alessandro Pastore, Roswitha Forstpointner, Simone Ferrero, Lothar Bergmann, Antonio Pezzutto, Oliver Weigert, Christian Scholz, Michael Unterhalt, Ulrich Keller, Marco Ladetto, Lorenz Truemper, Juergen Finke, and Martin Dreyling
- Subjects
Bendamustine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Follicular lymphoma ,Neutropenia ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,International Prognostic Index ,Internal medicine ,Multicenter trial ,Medicine ,business.industry ,Cell Biology ,Hematology ,medicine.disease ,3. Good health ,Surgery ,030220 oncology & carcinogenesis ,Radioimmunotherapy ,Rituximab ,Mantle cell lymphoma ,business ,030215 immunology ,medicine.drug - Abstract
Background Great efforts have been made in the last years to improve the therapeutical options of patients with mantle cell lymphoma (MCL), a distinct lymphoma subtype characterized by dismal long term prognosis. Although radioimmunotherapy (RIT) has been shown to be effective in follicular lymphoma, only one single-center experience of 31 evaluable patients has been published so far in relapsed MCL [Wang et al., JCO 2009]. The European MCL Network conducted a phase II, prospective, multicenter trial evaluating a single dose of yttrium-90-ibritumomab tiuxetan (90Y-IT) as reinduction or consolidation in patients with relapsed or refractory MCL. Patients and Methods Relapsed or refractory MCL patients after or not eligible for autologous stem cell transplantation (ASCT) with Results Between June 2004 and September 2008, 48 eligible patients were enrolled (16 group A, 32 group B) and 45 are evaluable for response. Median age was 68 years, 75% were males. Seventy-three % presented with high or intermediate risk MCL international prognostic index (MIPI), 42% with elevated lactate dehydrogenase, and 29% had bulky disease. Median number of previous therapies was 2 (range 1-5) in group A and 4 (range 1-6) in group B; 98% of patients received prior rituximab, 29% prior radiotherapy, 13% prior ASCT, 0% prior new agents and 15% had chemorefractory disease. The major toxicities consisted of myelosuppression, with thrombocytopenia in 21 patients (53%), neutropenia in 13 (33%) and anemia in 9 (23%; all grade 3/4, respectively), and one lethal bleeding. Non-hematologic grade 3/4 toxicities were gastrointestinal (n=3), infectious (n=1), and neurological (n=1), with a single patient (2%) developing a secondary myelodysplasia. Overall response rate (ORR) was 40% (20% CR) in group A and 72% (38% CR) in group B, with 5 patients converting from PR to CR. After a median follow-up of 38 months (range: 24-53 months), median PFS was 3.7 months in group A and 8.9 months in group B, translating in a median OS of 13.8 months and 31.2 months, respectively (Figure 1). In the 25 90Y-IT responders, PFS and OS were 23 months and 33.7 months, respectively, and patients responding to immunochemotherapy (group B) also showed a more favorable time to progression (16.9 months). No difference in survival rates was noticed according to MIPI, bulky disease, number and type of previous therapies, chemorefractoriness, and median time from initial diagnosis. Conclusions To the best of our knowledge, this is the largest trial assessing 90Y-IT therapy in relapsed/refractory MCL patients. Response rates in heavily pre-treated patients were comparable to those of other targeted approaches, though 90Y-IT showed a more favorable toxicity profile. Our experience suggests, that RIT applied as consolidation therapy might be the preferred approach, seeming to improve the quality and duration of response. Finally, responses appear to be independent from established risk factors and previous therapies. Further evaluation of the role of 90Y-IT in first-line therapy of MCL patients is ongoing. Disclosures: Off Label Use: Zevalin for treatment of relapsed mantle cell lymphoma. Scholz:Bayer: Speakers Bureau; Spectrum: Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau. Keller:Sepropharm: Consultancy. Dreyling:Jannsen: Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau, support of ITS, support of ITS Other; Celgene: Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau, support of ITS Other; Pfizer: Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau, support of ITS , support of ITS Other; Roche: Speakers Bureau, support of ITS , support of ITS Other; Mundipharma: support of ITS Other.
- Published
- 2013
41. Next-Generation Sequencing and Real-Time Quantitative PCR For Quantification Of Low-Level Minimal Residual Disease In Acute Lymphoblastic Leukemia Of Adults
- Author
-
Christiane Pott, Heiko Trautmann, Monika Brüggemann, Matthias Ritgen, Francois Pepin, Victoria Carlton, Dieter Hoelzer, Michael Kneba, Malek Faham, Marco Ladetto, and Nicola Gökbuget
- Subjects
Genetics ,Oncology ,Sanger sequencing ,medicine.medical_specialty ,Concordance ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Somatic evolution in cancer ,Minimal residual disease ,law.invention ,symbols.namesake ,law ,Acute lymphocytic leukemia ,Internal medicine ,Multiplex polymerase chain reaction ,medicine ,symbols ,Polymerase chain reaction - Abstract
Background Detection of minimal residual disease (MRD) in adult ALL is an independent risk parameter and early indicator of an impending relapse. MRD diagnostics have been incorporated in many ALL treatment protocols as a tool for pre-emptive treatment or risk group stratification. Currently, real-time quantitative (RQ)-PCR of immunoglobulin (IG) and T-cell receptor (TR) gene rearrangements is regarded as the most sensitive and standardized method to quantify MRD in BCR-ABL negative ALL. However, IG/TR oligoclonality and clonal evolution may lead to false negative results, while sensitivity varies depending on background amplification and is generally limited to 1E-05. Next-Generation Sequencing (NGS) of IG/TR gene rearrangements might overcome these limitations and potentially provide further increases in sensitivity, specificity, accuracy and reproducibility. We have performed a comparison of the two approaches on 130 diagnostic (DG), relapse (REL) and remission (FU) samples of 14 adult patients (pts) with B-cell precursor ALL. To test the NGS sensitivity, we partly selected follow-up samples for (low-level) RQ-PCR MRD positivity or a high pre-test probability of residual disease (either due to a later relapse or MRD detection in temporal relation to the analyzed sample). Patients and Methods 130 samples (97 bone marrow, 31 peripheral blood, 2 stem cell apheresis; 15 DG, 107 FU, 8 REL samples) were analyzed from 14 pts with B-cell precursor ALL. IG/TR-based RQ-PCR was carried out within routine diagnostics in the context of prospective clinical trials [Brüggemann et al., Blood 2006], according to the EuroMRD criteria [van der Velden et al., Leukemia 2007] at the MRD reference laboratory in Kiel. NGS was performed at the Sequenta facilities in South San Francisco. Using universal primer sets, IGH, IGK, TRB, TRG, and TRD variable, (diversity), and joining gene segments were amplified from DG and REL DNA. PCR products were sequenced to obtain a high degree of coverage and analyzed using standardized algorithms for clonotype determination. Tumor-specific clonotypes were identified for each patient based on their high-frequency in DG samples. The presence of the tumor-specific clonotype was then quantitated in FU samples. A quantitative and standardized measure of clone level among all leukocytes was determined using internal reference DNA. Comparability of MRD results by RQ-PCR and NGS was assessed using bivariate correlations between methods analysis (software R 3.0.1 with cor.test). Results Sanger sequencing of multiplex PCR detected 60 clonal IGH, IGK, TRB, TRG and/or TRD gene rearrangements at diagnosis (1-6/patient), NGS identified 57 index sequences (1-6/patient). Using NGS, relapse samples were analyzed for clonal evolution: 16/19 index rearrangements of relapsing pts were stable at relapse while 3/19 rearrangements (2 IGH VH-JH and 1 IGK) were lost at relapse. Based on Sanger sequence information of clonal rearrangements 1-3 RQ-PCR assays/patient were established while NGS focused on 1-4 clonal markers/patient. MRD was quantified in 107 FU in remission using both tools. A mean of 223,086±48,030 cells were analyzed per target using RQ-PCR, whereas NGS used 575,776±304,653 cells. An excellent concordance between the two methods was observed (p Conclusions Significant concordance was observed between NGS and RQ-PCR, with NGS having similar or slightly better sensitivity compared to standardized RQ-PCR. This can be partly attributed to the higher number of cells analyzed by NGS and points out that sensitivity advantages afforded by the NGS platform may be achieved by higher input cell amounts. Prospective analyses of unselected cases must be performed to verify the clinical impact of low level NGS-based MRD detection. Disclosures: Pepin: Sequenta Inc.: Employment, Stockholder Other. Carlton:Sequenta Inc.: Employment, Stockholder Other. Faham:Sequenta Inc.: Employment, Stockholder Other.
- Published
- 2013
42. Long-Term Molecular Results Of The Gimema VEL-03-096 Trial: Molecular Remission Achievement and Loss Are Major Outcome Predictors
- Author
-
Simone Ferrero, Marco Ladetto, Daniela Drandi, Federica Cavallo, Elisa Genuardi, Marina Urbano, Guglielmo Guarona, Vincenzo Callea, Clotilde Cangialosi, Tommaso Caravita, Claudia Crippa, Luca De Rosa, Antonietta Pia Falcone, Mariella Grasso, Tommasina Guglielmelli, Anna Marina Liberati, Francesco Pisani, Patrizia Pregno, Fausto Rossini, Stefania Oliva, Alberto Rocci, Roberto Passera, Mario Boccadoro, and Antonio Palumbo
- Subjects
medicine.medical_specialty ,Pathology ,Every Six Months ,business.industry ,Immunology ,Cell Biology ,Hematology ,Off-label use ,medicine.disease ,Biochemistry ,Minimal residual disease ,Thalidomide ,Transplantation ,Internal medicine ,medicine ,Lymphoid neoplasms ,business ,Survival analysis ,Multiple myeloma ,medicine.drug - Abstract
Background and aims The extensive use of new drugs in multiple myeloma (MM) allowed the achievement of unprecedented levels of cytoreduction and major advantages in survival rates, though almost all patients still relapse after a successful treatment. PCR-based minimal residual disease (MRD) studies are powerful prognostic tools, able to indentify patients at high risk of relapse. Thus, there is a growing interest in MRD to modulate therapy also in MM, as already happens in other lymphoid neoplasms. However available reports have a too short follow-up to be conclusive. In particular some points need to be addressed: 1) which is the long-term outcome of patients achieving molecular remission (MR) in the absence of further treatment? 2) What is the prognostic impact of MR loss? 3) How long is the window between MR loss and clinical relapse? These issues have been addressed based on the mature results of the GIMEMA VEL-03-096 trial [EudraCT Number 2004-000531-28], which currently has a median follow-up (mFU) of 93 months. Patients and methods Inclusion criteria and treatment schedule have been already reported [Ladetto et al., J Clin Oncol 2010]. MRD was assessed on bone marrow at diagnosis, study entry, after two VTD courses, at the end of treatment and then every six months up to clinical relapse. Patients underwent MRD detection using both qualitative nested PCR and Real Time Quantitative (RQ)-PCR, employing immunoglobulin heavy chain-derived patient specific primers, as described [Voena et al., Leukemia 1997; Ladetto et al., Biol Bone Marrow Transpl 2000; van der Velden et al., Leukemia 2007]. MR was defined as negative MRD results by nested-PCR or less than 1EE-04 by RQ-PCR. Loss of MR was defined as an increase of MRD levels of at least one log in consecutive samples at whenever timepoint. For survival analysis duration of response (DOR), progression-free survival (PFS), time to next treatment (TNT) and overall survival (OS) rates were used, as detailed in IMWG criteria [Rajkumar et al., Blood 2011]. Results Thirty-nine patients were enrolled. So far 27 serological progressions, 22 clinical relapses needing salvage treatment and 12 deaths (two non-MM-related) were observed. Median PFS was 60 months, median TNT 67 months and OS at mFU was 64%. 270 of the planned samples for MRD monitoring (86%) were actually received by the centralized lab. Currently, 26 MR and 11 MR losses have been registered. The achievement of MR was strongly associated with a better outcome, in terms of median DOR (62 vs 9 months, p Conclusions Besides confirming the strong prognostic value of PCR-based MRD monitoring in MM, our long-term results indicate the following: 1) the 42 months TNT of patients achieving MR underlines the excellent disease control of MM patients once obtained MR; 2) the occurrence of MR loss heralds relapse, with a TNT from MR loss comparable to TNT of patients not achieving MR; 3) there is a 9 months lag between MR loss and need for salvage treatment. These observations will have increasing relevance considering that ongoing methodological developments will allow effective MRD monitoring in the vast majority of MM patients. Disclosures: Off Label Use: Bortezomib and thalidomide as post-transplant consolidation during first-line treatment of multiple myeloma. Ladetto:Celgene: Research Funding, Speakers Bureau; Jannsen Cilag: Research Funding, Speakers Bureau; Mundipharma: Research Funding, Speakers Bureau; Roche: Research Funding, Speakers Bureau; Amgen: Research Funding, Speakers Bureau. Cavallo:Celgene: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Jannsen Cilag: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Research Funding. Caravita:Celgene: Honoraria, Research Funding; Jannsen Cilag: Honoraria. Guglielmelli:Celgene: Research Funding. Boccadoro:Celgene: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Research Funding; Jannsen Cilag: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Research Funding. Palumbo:Bristol-Myers Squibb: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; Janssen Pharmaceuticals: Consultancy, Honoraria; Millenium: Consultancy, Honoraria; Onyx: Consultancy, Honoraria; Amgen: Consultancy, Honoraria.
- Published
- 2013
43. Relapsed Or Refractory Aggressive Non Hodgkin B-Cell Lymphomas Treated With Lenalidomide With/Without Rituximab Or Steroids: A Single Center Restrospective Study
- Author
-
Patrizia Pregno, Maura Nicolosi, Alessia Castellino, Barbara Botto, Umberto Vitolo, Paola Riccomagno, Manuela Ceccarelli, Daniele Caracciolo, Paola Ghione, Annalisa Chiappella, Giulia Benevolo, Chiara Ciochetto, Lorella Orsucci, Manuela Zanni, Marco Ladetto, and Clara Pecoraro
- Subjects
medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Gastroenterology ,Surgery ,Transplantation ,International Prognostic Index ,Internal medicine ,medicine ,Mantle cell lymphoma ,Rituximab ,business ,Diffuse large B-cell lymphoma ,Dexamethasone ,Lenalidomide ,medicine.drug - Abstract
Introduction Patients with relapsed or refractory aggressive non Hodgkin B-cell lymphomas (NHL) not eligible to high dose chemotherapy and transplantation had a dismal prognosis. Lenalidomide showed activity in this setting as single agent or in combination. On this basis, we conducted a single center retrospective study to investigate efficacy and safety of lenalidomide alone or in association with rituximab or steroids in patients with heavily pretreated aggressive B-cell NHL. Methods Primary end points of the study were response rate (RR), defined as complete response (CR), partial response (PR) and stable disease (SD), and duration of response (DOR); secondary end points were feasibility and safety. Inclusion criteria for the analysis were: patients with relapsed/refractory aggressive B-cell NHL, aged >18 years, treated with lenalidomide between August 2007 to June 2012. Treatment scheme were: standard dose of oral lenalidomide 25 mg/day for 21 days every 28 days as single agent; standard dose of lenalidomide with the same schedule in association to weekly dexamethasone (20 mg bolus); lenalidomide 20 mg/day for 21 days every 28 days in combination with rituximab (375 mg/sqm) every 28 days. Patients were treated until disease progression or unacceptable toxicities. Results A total of 53 patients were analyzed. Different histotypes of NHL were included in the study: 34 diffuse large B-cell lymphomas (DLBCL), 11 mantle cell, 5 follicular, 2 primitive mediastinal B-cell and one Burkitt lymphoma. At relapsed before lenalidomide treatment, the majority of the patients presented an advanced disease: 40 (75%) stage 3-4; intermediate high/high risk international prognostic index (IPI) 23 (43%). Bone marrow was involved in 20 (38%) patients and 20 (38%) cases presented a bulky disease. Prior treatment lines were as follows: 8 (15%) patients received lenalidomide at first relapse while 24 (45%) underwent more than 3 previous lines of therapy; 14 (26%) patients received high dose chemotherapy and autologous stems cell transplant, one (2%) patient was allogenic transplanted and 4 (8%) did both before lenalidomide. All patients analyzed received lenalidomide: 31 patients (58%) underwent lenalidomide as single agent, 11 (21%) received a combination scheme of lenalidomide plus rituximab and 11 cases (21%) were treated with lenalidomide plus steroids. Median time from diagnosis and the beginning of lenalidomide was 25,3 months (3,7- 145,9), while median time from last previous therapy and lenalidomide treatment was 3,2 months (0,4- 38). At the time of this analysis response assessment was done in 51 patients: Response rate was 35% (18 patients), with CR 8 (15%), PR 5 (10%), SD 5 (10%). All patients who obtained CR underwent more than 3 courses of therapy, while among 31 patients who did not respond to treatment, 21 failures occurred during the first three cycles. Concerning different schemes of therapy: in the arm treated with lenalidomide as single agent RR was 24%, among patients underwent lenalidomide plus rituximab was 55% and in the group receiving lenalidomide plus steroid 45%. Among 34 DLBCL patients, RR was 41% (n= 14: CR 5, PR 4, SD 5), while in 11 patients affected by mantle cell lymphoma RR was 27% (n= 3: CR2, PR 1). Median DOR for all 18 responding patients was 12 months (0,2-24). At a median follow-up of 20 months, 5 patients were in stable CR, 7 continued lenalidomide, 11 relapsed and 28 died. Patients received a total of 257 cycles of lenalidomide, of which 25 were earlier interrupted and 48 were reduced in dose or duration; 50% of patients had at least one interruption in the planned treatment, however globally 91% of the expected dose was given. One patient died due to heart failure during the treatment. Toxicity was globally mild: most common grade 3-4 adverse events were neutropenia (19%), anemia (17%) and thrombocytopenia (17%). Five patients had grade 3-4 infections and 3 patients had thromboembolic events (only one grade 3). Two cases of neuropathies, both grade ≤ 2 were observed. Conclusions Lenalidomide single agent or in association with rituximab or steroids is effective and safe in patients with relapsed or refractory aggressive B-cell NHL, showing a promising response rate also in patients with heavily pretreated disease and with a mild toxicity profile. Disclosures: Vitolo: Roche: Speakers Bureau; Celgene: Speakers Bureau; Takeda: Speakers Bureau.
- Published
- 2013
44. Improved Igh-Based MRD Detection By Using Droplet Digital PCR: a Comparison With Real Time Quantitative PCR In MCL and MM
- Author
-
Daniela Drandi, Lenka Kubiczkovà, Nadia Dani, Simone Ferrero, Luigia Monitillo, Barbara Mantoan, Elisa Genuardi, Daniela Barbero, Manuela Gambella, Davide Barberio, Paola Ghione, Guglielmo Guarona, Elona Saraci, Paola Omedè, Roberto Passera, Roman Hajek, Sergio Cortelazzo, Antonio Palumbo, Mario Boccadoro, Giorgio Inghirami, and Marco Ladetto
- Subjects
Pathology ,medicine.medical_specialty ,Thermal cycler ,Serial dilution ,business.industry ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,Gene rearrangement ,030204 cardiovascular system & hematology ,Biochemistry ,Minimal residual disease ,Molecular biology ,3. Good health ,Standard curve ,03 medical and health sciences ,0302 clinical medicine ,Real-time polymerase chain reaction ,Medicine ,Digital polymerase chain reaction ,business ,030215 immunology - Abstract
Background In mature lymphoid disorders, minimal residual disease (MRD) detection based on real time quantitative PCR (RQ-PCR) of immunoglobulin heavy chain gene rearrangement (IgH) has a well-established role in prognostic assessment, particularly in Mantle cell Lymphoma (MCL) and Multiple Myeloma (MM). RQ-PCR has excellent sensitivity and specificity but has a major limitation in its relative quantification nature, as it requires a reference standard curve usually built with dilutions of diagnostic tumor DNA or on plasmids containing the target rearrangement. Droplet Digital PCR (DD-PCR), applying the principle of limiting dilution of DNA and single molecule detection allows a reliable absolute quantification of target. In this study we compared IgH-based MRD detection by RQ-PCR and DD-PCR, to assess whether DD-PCR could achieve the same performances of RQ-PCR in the absence of the limitation mentioned above. Methods Bone marrow (BM) and peripheral blood (PB) samples were collected from patients affected by MCL and MM in which RQ-PCR based MRD analysis was already performed in the context of prospective clinical trials. In all trials patients gave the informed consent for MRD determination. IgH-based MRD detection by RQ-PCR was carried out as previously described [Ladetto et al. BBMT 2000] and results were interpreted according to the Euro-MRD guidelines [van der Velden et al. Leukemia 2007]. DD-PCR was performed by the QX100 Droplet Digital PCR system (Bio-RAD Inc.) on 500 ng of genomic DNA combined with the same Allele Specific Oligonucleotides (ASO)-primers and TaqMan-probes used in the RQ-PCR. Droplets were generated by QX100 droplet generator. End-point PCR (40 cycles) was performed on a T100 Thermal cycler (Bio-RAD Inc). The PCR product was loaded in the QX100 droplet reader and analyzed by QuantaSoft 1.2 (Bio-Rad Inc). For data interpretation RQ-PCR and DD-PCR results were expressed as amount of target copies per 1E+05 cells. Comparability of MRD results by DD-PCR and RQ-PCR was assessed by means of bivariate correlations between methods analysis (R2.15.1 package irr). Discordances were classified as follows: a positive/negative discordance was defined as major when the positive result was >1E-04 and minor when ≤1E-04; a quantitative discordance was defined as the presence of two positive results with a quantitative discrepancy >1 log. Results Overall, 161 samples belonging to 35 patients (18 MCL and 17 MM), 66 MCL and 95 MM were analyzed. 35 samples were taken at diagnosis and 126 at follow-up. 118 were BM while 43 were PB. A significant correlation was found between DD-PCR and RQ-PCR (R2=0.89, p Conclusions Here we report for the first time the use of DD-PCR in the context of IgH-based MRD evaluation in lymphoproliferative disorders. DD-PCR is a feasible tool for IGH-based MRD monitoring in MCL and MM, reaching similar sensitivities compared to standardized RQ-PCR. Moreover DD-PCR allows bypassing the need of building a standard curve thus considerably reducing the complexity of IgH-based RQ-PCR (need of purified diagnostic tissue or Flow Cytometry-based quantification of tumor load or diagnosis, or building of a plasmid-derived standard curve). Finally DD-PCR might potentially overcome the problem of positive non-quantifiable samples. These features make DD-PCR a feasible and attractive alternative method for IgH-based MRD assessment. Disclosures: Kubiczkovà: GAP304/10/1395 : Research Funding; MUNI/11/InGA17/2012: Research Funding.
- Published
- 2013
45. Prolonged Survival Of Poor Risk Follicular Lymphoma Patients Following Primary Treatment With Rituximab-Supplemented CHOP Or HDS With Autograft: Long-Term Results Of The Multicenter Randomized GITMO/FIL Trial
- Author
-
Corrado Tarella, Fabio Benedetti, Carola Boccomini, Caterina Patti, Anna Maria Barbui, Alessandro Pulsoni, Maurizio Musso, Anna Marina Liberati, Guido Gini, Claudia Castellino, Fausto Rossini, Fabio Ciceri, Delia Rota Scalabrini, Caterina Stelitano, Francesco Di Raimondo, Tommasina Perrone, Alessandra Tucci, Atto Billio, Francesco Zallio, Valerio Zoli, Angela Congiu, Franco Narni, Alessandra Dondi, Guido Parvis, Gianpietro Semenzato, Paolo Corradini, Riccardo Bruna, Angela Gueli, Barbara Mantoan, Roberto Passera, Michele Magni, and Marco Ladetto
- Subjects
medicine.medical_specialty ,Intention-to-treat analysis ,business.industry ,Immunology ,Late effect ,Follicular lymphoma ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Gastroenterology ,Minimal residual disease ,Surgery ,Tolerability ,Prednisone ,Internal medicine ,Clinical endpoint ,Medicine ,Rituximab ,medicine.symptom ,business ,medicine.drug - Abstract
Introduction A randomized multicenter study of 134 Follicular Lymphoma (FL) patients, selected for age less than 60 yrs. and poor prognostic features according to age-adjusted IPI (2-3) and IIL-score (3 or greater) was conducted between March 2000 and May 2005, among 30 Italian Centers. The study compared efficacy and tolerability of CHOP-R vs. R-HDS with autograft as primary treatment in poor-risk FL. Initial results have been already reported (Ladetto M et al, Blood 2008), showing superior disease control with R-HDS without any survival advantage. We have recently updated the long-term outcome and the results at long-term are here presented at a median follow-up of 9.5 yrs. Patients and Methods Of the original 134 randomized patients, the long-term outcome has been updated for 125 patients, 61 of CHOP-R and 64 of R-HDS arms. Clinical characteristics at study entry and treatment schedules have been already reported. Briefly, the main features of the updated patients included: median age 51 yrs. (22-60), M/F ratio 74/51, aaIPI 2-3 90%, FLIPI >2 (retrospectively assigned) 60%, high LDH 49%, bulky disease 62%, B-symptoms 45%, BM involvement 86%. Clinical characteristics were balanced among the two arms. Treatment schedule consisted of: i. standard arm: 6 courses of cyclo-phosphamide/doxorubicin/vincristine/prednisone followed by 4-weekly rituximab courses (CHOP-R); ii. experimental arm: rituximab-supplemented high-dose sequential chemotherapy with autografting (R-HDS). The analysis was intention to treat with event-free survival as the primary endpoint. Minimal residual disease was evaluated post treatment in 58 patients with a bcl-2/IgH MBR or mcr translocation confirmed at diagnosis by nested PCR. The trial was registered at www.clinicaltrials.gov as no. NCT00435955. The long-term outcome has been updated in July 2013 by 28 out of 30 participating Centers accounting for 125 patients (93% of the whole series). Results Complete remission (CR) was achieved by 88 (70.4%) patients, including 35 (57%) with CHOP-R and 53 (83%) with R-HDS (p < .001); in addition, 37 out of 58 (64%) patients achieved a Molecular Remission (MR). At a median follow-up (MFU) of 9.5 yrs., 88 patients (70.4%) are alive. Overall, 19 patients died for lymphoma progression (11 in the CHOP-R, 8 in the R-HDS arms), there were nine deaths for secondary malignancy (3 in the CHOP-R, 6 in the R-HDS arms), nine more patients died for other causes, including four early toxic deaths. The overall survival projection for the whole series is 78% and 70% at 5 and 10 yrs., respectively. As shown in Figure 1, there were no main differences in the long-term OS between the two arms, with 5 and 10 yrs projections respectively of 75% and 70% for CHOP-R and 81% and 70% for R-HDS (p=0.96). Response to primary treatment had a major impact on the OS, with 5 and 10 yr survival projections respectively of 90% and 80% for patients achieving CR, and of 49 and 43 for those with less than CR (p < .001) (Figure 2A). Similarly, MR achievement was associated with prolonged overall survival, with 5 and 10 yr survival projections respectively of 89% and 83% for patients with PCR-ve on BM cells, and of 76 and 57 for those with persistent PCR-positivity (p = .03) (Figure 2B). Conclusion The long-term follow-up of the randomized CHOP-R vs. R-HDS trial indicate that: i. poor risk FL may now experience a prolonged survival, with approximately 70% of patients alive at 10 yrs., due to the combined efficacy of both primary chemo-immunotherapy and salvage treatments; ii. the superior disease control of R-HDS compared to CHOP-R does not translate in any survival advantage, with analogous OS regardless of which treatment is used; iii. also in FL like in other lymphoproliferative malignancies, achieving CR and MR is crucial not only for the disease control but also for long-term overall survival; iv. lymphoma progression remains the major cause of death, while secondary neoplasms, in particular secondary leukemias represent the second cause of treatment failure. Thus, efforts are still needed in order to increase the anti-tumor efficacy while reducing any potential late effect in treatment options for FL. Disclosures: Tarella: Roche Co.: support and honoraria for Conference participation Other. Ladetto:Roche: Honoraria, Research Funding, Speakers Bureau.
- Published
- 2013
46. PCR-Based Minimal Residual Disease (MRD) Detection Is a Strong Independent Outcome Predictor Also in Rituximab-Intensive Non-ASCT-Based Programs: Results From the ML17638 Multicenter Randomised Phase III Trial for Elderly Follicular Lymphoma (FL) Patients of the Fondazione Italiana Linfomi (FIL)
- Author
-
Pellegrino Musto, Luigi Rigacci, Renato Fanin, Giorgina Specchia, Chiara Lobetti-Bodoni, Giuseppe Rossi, Enrica Gamba, Elisa Genuardi, Giovannino Ciccone, Guido Parvis, Francesco Di Raimondo, Antonello Pinto, Alessandro Pulsoni, Mario Petrini, Andrea Evangelista, Delia Rota-Scalabrini, Barbara Botto, Barbara Olivero, Barbara Mantoan, Carola Boccomini, Marco Ladetto, Luca Baldini, Luigi Gugliotta, Umberto Vitolo, Andrea Gallamini, and Stefano Sacchi
- Subjects
medicine.medical_specialty ,Randomization ,Proportional hazards model ,business.industry ,Immunology ,Follicular lymphoma ,Context (language use) ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Minimal residual disease ,Monitoring program ,Surgery ,Fludarabine ,Internal medicine ,medicine ,Rituximab ,business ,medicine.drug - Abstract
Abstract 787 Background: Many studies support the value of PCR-based MRD detection using the bcl-2-IgH translocation as an outcome predictor in FL but some failed to confirm this observation. Concerns have been raised particularly for programs which are highly Rituximab (Rtx) intensive (with or without maintenance) and non-ASCT-based. The ML17638 study, contained an extensive centralized MRD monitoring program, whose results are here presented. Patients and methods: Clinical results of study have been already reported (Vitolo et al, ASH 2011). The program consisted of 4 R-FND courses (Rtx, fludarabine, mitoxantrone, dexamethasone) followed by 4 doses of weekly Rtx. Patients (pts) achieving 3partial response (PR) were randomized to Rtx maintenance (arm A) or observation (arm B). A total of 234 untreated elderly (age 60–75 years) pts at diagnosis were enrolled. With a median follow-up from randomization of 34 months, 3-year PFS and OS were 66% (95%CI:59-72%) and 89% (95%CI:85-93%), with a clear trend in favor of arm A for 2-year PFS (81% vs 69%). At enrolment, pts were screened for a molecular marker based on the bcl-2/IgH MBR or mcr. If found, pts were tested at 8 fixed timepoints: at month 5 (M5) after 4-R-FND, at the end of induction therapy (M8) and during maintenance/observation and follow-up (M12,M18,M24,M30,M36 and M42) or until relapse. MRD was assessed by both nested PCR (n-PCR) and real time quantitative PCR (RQ-PCR) on BM cells. Methods have been already reported (Ladetto Exp Hematol 2001). RQ-PCR was performed and analyzed according to the Euro-MRD guidelines (Van der Velden Leukemia 2007). The lab performs routine quality controls in the context of Euro-MRD and was blinded to clinical results and radomization arm. The impact of MRD on PFS was evaluated by log-rank tests and Cox models including age, sex, FLIPI, ECOG PS and complete remission (CR). In addition, the effect of PCR negativity on PFS during the whole follow-up period was evaluated by a time-varying covariate included in the models, also considered in a cumulative way (0, 1, 2, 3 or more consecutive PCR-negative timepoints). Results: 229 of 234 enrolled pts (98%) were screened at study entry. A molecular marker was found in 118 (51.5%). Of these, 9 were excluded due to withdrawal before M5 (7) or inadequate sampling (2). Overall, 800 follow-up samples were expected. Of these, 707 (88%) were received and analysed: 98% of pts were evaluable for 350% of timepoints and 87% for 375%. Pts with and without a marker had identical PFS (61% at 42m for both). Sixty six per cent of pts achieved PCR-negativity after R-FND and 81% at the end of treatment, with a mean tumor burden reduction of 11 natural logaritm after R-FND and a further decrease of 1.6 after the 4 weekly Rtx. At randomization, PCR-positivity rate was similar in the two arms while during and after maintenance pts in Arm A had a lower rate of PCR-positivity (9% vs 17% p=0.02). The achievement of PCR-negativity by both n-PCR and RQ-PCR at timepoints M8,12,18 and 24 predicted a better PFS (M5 not predictive, M30, 36, 42 have too early follow up for meaningful evaluation). After M8, 2-year PFS was significantly better in PCR-negative than PCR-positive pts: 72% vs 39% (p=0.007, Fig. 1). Achieving a double PCR-negativity at M8-M12 or triple molecular negativity at M8-M12-M18 was associated with a further increase of PFS (82% vs 46% for months 8–12, p=0.001 and 87% vs 53% for months 8–12-18, p=0.001). PCR-negativity at M8 ensured a subsequent better PFS both in CR (p=0.023, HR=0.33, 95%CI: 0.13–0.86) and PR (p=0.074, HR=0.28, 95%CI: 0.07–1.13) pts (Fig. 2). Disclosures: Ladetto: Hoffman-La Roche: Consultancy, Honoraria. Rossi:Roche: Honoraria. Musto:Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Gamba:Roche: Employment. Vitolo:Celgene: Honoraria; Janssin-Cilag: Honoraria; Roche: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2012
47. Large Genomic Aberrations Are Independent Prognosticators of A Shorter Time to First Treatment (TTT) in Chronic Lymphocytic Leukemia (CLL) Patients with A Normal FISH
- Author
-
Mario Uhr, Francesco Forconi, Emanuele Zucca, Georg Stussi, Andrea Rinaldi, Francesco Bertoni, Michael Mian, Franco Cavalli, Ivo Kwee, Roberto Marasca, Gianluca Gaidano, Afua Adjeiwaa Mensah, Davide Rossi, and Marco Ladetto
- Subjects
Oncology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Chronic lymphocytic leukemia ,Concordance ,Immunology ,Single-nucleotide polymorphism ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,genomic DNA ,Leukemia ,Internal medicine ,medicine ,IGHV@ ,Trisomy ,business ,Fluorescence in situ hybridization - Abstract
Abstract 3906 The clinical course of CLL patients ranges from an indolent and chronic disease to a rapidly progressing leukemia or lymphoma necessitating aggressive treatment. Very recent data have shown that genomic complexity evaluated by conventional karyotyping with DSP30/IL2 stimulation (Rigolin et al, Blood 2012) can be helpful in the prediction of the clinical course of patients with a normal fluorescence in situ hybridization (FISH) panel according to Dšhner et al (NEJM 2000). However, large studies investigating the impact of genomic complexity on the clinical course and possible correlations with other clinical parameters at time of diagnosis are still lacking. Therefore, we analyzed the role of genomic complexity in a large series of 329 CLL patients with available clinical data. Also, since SNP-array might soon represent an alternative to standard FISH in the clinical routine, we performed the so far largest comparison of FISH versus SNP-array in CLL. Methods. Copy-number data, obtained using the Affymetrix Human Mapping GeneChip 6.0 arrays (SNP6), were derived from our previously reported CLL dataset (Rinaldi, Mian, Kwee et al, BJH 2011). The FISH panel interrogated deletions at 13q14.3, 11q22, 17p13 and trisomy 12. CLL diagnosis and management were based on the NCI Working group criteria (Hallek et al., Blood 2008). Real-time PCR on genomic DNA was performed to validate discordant FISH/SNP6 results. Results. Seventy-seven of the 329 CLL patients (23%) presented a normal FISH. Among those, 17 patients (22%) had at least one large (>5 Mb) genomic aberration, different from those described by Dšhner et al (NEJM 2000). There was no correlation with the presence of TP53 mutations, since 13/13 were wtTP53. The DNA gains or losses did not occur at specific genomic loci, and their presence significantly affected the TTT (P= 0.0010; R=2.8; 95% CI, 1.5–5.5) (Figure 1), but not overall survival (OS) (p= 0.098; HR 2.3; 95% CI, 0.83–6.6). In a multivariate analysis including age, Binet stage, IGHV genes mutational status and the large genomic lesions, the latter three factors emerged as independent prognosticators for TTT with a significance of P The concordance between FISH and SNP-array results was 93% for all analyzed genomic regions: 97% for trisomy 12, 96% for del 11q, 95% for del 17p and 84% for del 13q. False positive FISH results were observed for del 13q, while among the 18 cases with 17p loss by FISH half of the 10 cases with less than 40% of nuclei carrying the lesion were classified as normal by SNP6. We applied the FISH-based prognostic model developed by Dšhner et al (NEJM 2000) to our cohort classifying the 329 patients using results obtained by FISH or by SNP6: the Kaplan-Meier curves were comparable between FISH and SNP-array, both for OS as for TTT, and the log-rank test was highly significant for both approaches. Conclusions. Patients with a normal FISH but with large genomic lesions detected by SNP-array have a worse TTT. As a whole, SNP-array has a high sensitivity and specificity and is able to identify the most important known prognostic genomic aberrations of CLL. A validation in prospective trials is needed. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
48. The Peripheral Blood Lymphocyte to Monocyte Ratio At Diagnosis Is a Potent Outcome Predictor in Diffuse Large B-Cell Lymphoma Treated with R-CHOP: A Long-Term Analysis On 973 Patients Receiving Chemotherapy with or without Rituximab
- Author
-
Anna Maria Barbui, Federica Delaini, Cristina Boschini, Marco Ladetto, Daniele Caracciolo, Angela Gueli, Alessandro M. Gianni, Alessandro Rambaldi, Andrea Rossi, Elena Oldani, Corrado Tarella, Liliana Devizzi, Giuseppe Gritti, Roberto Passera, and Alberto De Crescenzo
- Subjects
Univariate analysis ,medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,CHOP ,medicine.disease ,Biochemistry ,Gastroenterology ,Surgery ,Log-rank test ,symbols.namesake ,International Prognostic Index ,Median follow-up ,Internal medicine ,medicine ,symbols ,Rituximab ,business ,Diffuse large B-cell lymphoma ,Fisher's exact test ,medicine.drug - Abstract
Abstract 1553 Background and Aim The number and type of lymphocytes and monocytes/macrophages detectable in the peripheral blood and the lymph nodes of patients with Hodgkin and non-Hodgkin lymphomas has been recently extensively investigated and interesting results indicate they may possibly affect the pathogenesis and prognosis of these diseases. Recent results indicate that the lymphocyte/monocyte ratio (LMR), when assessed at diagnosis by a simple automatic blood count, may predict the clinical outcome of diffuse large B cell lymphoma patients (DLBCL) treated with the R-CHOP chemotherapy program (Li Z-M et al.: PLoS ONE 7(7):2012). The main objective of our study was to evaluate whether: a) the prognostic value of LMR could be confirmed either in patients treated with CHOP and Rituximab (R-CHOP) as well as in those treated with CHOP alone and b) the LMR could improve the prognostic profile as defined by the International Prognostic Index (IPI). Patients and Methods We retrospectively reviewed the clinical outcome of 973 DLBCL patients treated (549 with R-CHOP) and regularly followed at our institutions from 1984 to 2012. The median age of this patients cohort was 61 years (range, 18–86), the Male/Female ratio 55% and the median follow up 44 months (range, 2 – 330). According to the IPI score, 61% of patients were in the low (0–2) risk group, while 39% were in the intermediate or high-risk groups (3–5). A receiver operating characteristic (ROC) curve analysis was used to illustrate in our data set the best cut off values of peripheral blood lymphocyte absolute count (ALC), monocyte absolute count (AMC) or LMR. The relationship between IPI and the LMR was analyzed by the Fisher exact test. Univariate analysis to evaluate differences between variables was performed by the log rank. A multivariate analysis was performed by Cox proportional-hazards models. Results A preliminary ROC curve analysis performed on all patients (treated with or without rituximab) failed to identify any meaningful relationship between Overall Survival (OS) and the ALC, the AMC or the LMR. However, when the same analysis was restricted to patients treated with R-CHOP, we could confirm not only the positive correlation between ALC and AMC and OS, but most importantly that a LMR value >2.6 is the most sensitive (70%) and specific (53%) cut off to predict the OS. Within the R-CHOP treated cohort (N=549), we further investigated the relationships between LMR and the most relevant clinical features measured at diagnosis. Patients with a LMR ≤ 2.6 (52%) had a worst ECOG PS (p= 0.000), a higher LDH level (P= 0.000), a higher IPI (p= 0.000) and more frequently they were male (p= 0.02) and had an advanced Ann Arbor disease stage (p= 0.002). On the contrary, no statistical correlation was observed with age and the presence of extranodal sites. The proportion of patients achieving a complete response or a very good partial response was 95% in patients with a LMR >2.6 and 87% for those with a LMR ≤ 2.6 (p=0.018). More interestingly, among patients failing to achieve CR the proportion of those with a LMR ≤ 2.6 was 79% as compared to 21% among those with a LMR >2.6. The Event Free Survival of patients with a LMR >2.6 was significantly better when compared to those with a LMR ≤ 2.6 (71% vs. 59% at 5 years, p= 0.01) while no difference was observed for the Disease Free Survival. By univariate and multivariate analysis we could show that, similarly to a high IPI, a LMR ≤ 2.6 strongly predict a poor OS (p= 0.0000) (Figure 1). In addition, we could demonstrate a strong interaction between IPI and LMR since patients with a high IPI and LMR ≤ 2.6 are characterized by a very poor prognosis when compared to all the others (p= 0.000). Conclusions Our results confirm that a LMR ≤ 2.6 when assessed at diagnosis by a simple automatic blood count is not only a strong predictor of poor survival but it may help to better define a very poor prognostic subgroup in R-CHOP treated DLBCL. This novel prognostic marker is irrelevant when applied to patients receiving chemotherapy alone, giving further support to the notion that lymphocytes and/or monocytes play a crucial role on the therapeutic activity of Rituximab. Disclosures: Rambaldi: Hoffman-La Roche: Consultancy, Honoraria. Ladetto:Hoffman-La Roche: Consultancy, Honoraria. Gianni:Hoffman-La Roche: Consultancy, Honoraria. Tarella:Hoffman-La Roche: Consultancy, Honoraria.
- Published
- 2012
49. High Rates of Prolonged Molecular Remissions After Tandem Autologous-Nonmyeloablative Allografting in Newly Diagnosed Myeloma
- Author
-
Paola Omedè, Moreno Festuccia, Roberto Passera, Nicola Mordini, Renato Fanin, Sara Barbiero, Silvia Cena, Simone Ferrero, Benedetto Bruno, Luisa Giaccone, Francesca Patriarca, Daniela Barbero, Luigia Monitillo, Antonio Palumbo, Alberto Rocci, Marco Ladetto, Andrea Gallamini, Mario Boccadoro, and Daniela Drandi
- Subjects
Melphalan ,Immunofixation ,medicine.medical_specialty ,biology ,business.industry ,Immunology ,Cell Biology ,Hematology ,Gene rearrangement ,Total body irradiation ,medicine.disease ,Biochemistry ,Gastroenterology ,Minimal residual disease ,Surgery ,Leukemia ,medicine.anatomical_structure ,Internal medicine ,biology.protein ,Medicine ,Bone marrow ,business ,Multiple myeloma ,medicine.drug - Abstract
Abstract 4204 Background and aims. Myeloablative allografting induces high rates of persistent molecular remissions (MR) in multiple myeloma (MM) [Corradini et al, J Clin Oncol 1999] and greatly reduces the risk of relapse. Similar results have also been reported after reduced-intensity conditioning [Kröger et al, Blood abstr.2011]. Long term data on minimal residual disease (MRD) kinetics after tandem autologous-nonmyeloablative allografting (auto-allo) are lacking. We here present the results of MRD analyses by nested qualitative PCR and real time quantitative (RQ) PCR on a series of 26 newly diagnosed MM patients treated with auto-allo on a prospective clinical trial [ClinicalTrial.gov, NCT-00702247, Bruno et al, Blood 2009]. Patients and methods. Between 1999 and 2009, 19/26 (73%) stage II-III MM patients, median age 52 years (range 42–65), who had a diagnostic bone marrow (BM) specimen suitable for immunoglobulin heavy-chain gene rearrangement (IGH) sequencing were evaluated for MRD by PCR-based methods. The tandem approach consisted of an autograft with melphalan, 200 mg/m2, followed, upon recovery, by non-myeloablative 200 cGy total body irradiation and allogeneic peripheral stem cell infusion. Post-grafting immunosuppression consisted of mycophenolate mofetil and cyclosporine. BM samples were collected at diagnosis, after autograft, at month 1, 3, 6 after the allograft and then every 6 months. Nested-PCR and RQ-PCR analyses were carried out using IGH patient-specific primers, as previously described [Voena et al, Leukemia 1997; Ladetto et al, Biol Bone Marrow Transpl 2000]. For outcome analysis patients were grouped according to reported criteria [Ladetto et al, Blood abstr.2011]. Briefly, FullMR and StandardMR indicated MRD negativity on two consecutive samples by respectively nested-PCR or RQ-PCR (less sensitive but better standardized, according to European Study Group on MRD detection guidelines [van der Vendel et al, Leukemia 2007]). Clinical complete remission (CCR) required absence of serum and urine M-component by electrophoresis and immunofixation and less than 5% plasma cell infiltration in BM aspirates by flow-cytometry. Results. In 19 of 26 patients (73%) a molecular marker was found. In these cases at a median follow-up of 10 years (4.4–12) from diagnosis and 8.9 years (3.5–11) from the allograft, overall survival (OS) was 61% and median time-to-progression (TTP) 5.6 years. Overall, transplant-related mortality occurred in 3/19 patients (16%), while four out of 19 patients (21%) died of disease progression. MRD studies carried out on a total of 148 BM samples showed that after the autograft 3/19 patients (16%) were negative by nested-PCR. After the allograft the rates of PCR-negativity gradually increased up to 4/18 (22%) at 1 and 3 months, 7/17 (41%) at 6 months and 8/15 (53%) at 1 year post-transplant. Overall, 8 patients achieved FullMR at a median time from allograft of 6 months (1–12) and for a median duration of 33 months (6–102). Overall, 8 relapses were observed, 6 in the 11 patients who never achieved FullMR and 2 in patients who reached FullMR: of these one showed a molecular relapse with persistent CCR after sustained FullMR of 3 years and the other relapsed 6 months after last PCR-negative sample. Patients in FullMR had a significantly better median TTP (not reached vs 1.6 years, p=0.043) and OS (not reached vs 3.3 years, p=0.008) than patients who did not achieve FullMR (Fig. 1). StandardMR occurred in 12/19 patients (63%) during the first 24 months post-transplant, at a median time of 2 months (1–18) and for a median duration of 27 months (3–102). Patients in StandardMR showed a significantly better median TTP (not reached vs 1 year, p=0.005) and OS (not reached vs 3.3 years, p=0.031) as compared to patients who not achieved StandardMR (Fig. 2). All patients in either FullMR or StandardMR were also in CCR. There was no significant correlation between occurrence of chronic graft-vs-host disease and MR, suggesting specific graft-vs-myeloma effects. Conclusions. Auto-allo approach in newly diagnosed MM induces high rates of prolonged FullMR and StandardMR (up to 50%), similar to those reported after myeloablative allografting. MR significantly associated with better TTP and OS and increased during the first year post-allografting, clearly documenting the existence of an effective and persistent graft-vs-myeloma effect, potentially curative in a subset of patients. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
50. Next-Generation Sequencing and Real-Time Quantitative PCR for Minimal Residual Disease (MRD) Detection Using the Immunoglobulin Heavy Chain Variable Region: A Methodical Comparison in Acute Lymphoblastic Leukemia (ALL), Mantle Cell Lymphoma (MCL) and Multiple Myeloma (MM)
- Author
-
Malek Faham, Francois Pepin, Daniela Drandi, Marco Ladetto, Victoria Carlton, Mario Boccadoro, Antonio Palumbo, Luigia Monitillo, Roberto Passera, Monika Brüggemann, Christiane Pott, Simone Ferrero, and Heiko Trautmann
- Subjects
Oncology ,Pathology ,medicine.medical_specialty ,business.industry ,Concordance ,Immunology ,Context (language use) ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Minimal residual disease ,Somatic evolution in cancer ,law.invention ,law ,Acute lymphocytic leukemia ,Internal medicine ,medicine ,Mantle cell lymphoma ,business ,Polymerase chain reaction ,Multiple myeloma - Abstract
Abstract 788 Background. Real-Time Quantitative (RQ) PCR-based MRD detection using tumor-specific primers derived from the immunoglobulin heavy chain variable region (IGH) is an established disease monitoring tool with high predictive value in ALL, MCL and MM. It is highly sensitive and has been standardized in the context of international cooperative groups such as the European Scientific Foundation for Laboratory Hematooncology (ESLHO). However it has some limitations, including marker identification failure, particularly in hypermutated tumors and false negatives due to clonal evolution, particularly in ALL. IGH-based next-generation sequencing (NGS) might overcome some of these limitations and further increase sensitivity, specificity, accuracy and reproducibility. We have thus performed a head to head comparison of the two methods on diagnostic (DG) and post-treatment follow-up (FU) samples on a panel of 55 patients. Patients and Methods. Overall, 381 samples (215 bone marrow, 166 peripheral blood; 62 DG, 319 FU) were collected from 55 patients (15 ALL, 30 MCL, 10 MM) in which RQ-PCR based MRD analysis had been performed in the context of prospective clinical trials. IGH-based RQ-PCR was carried out as previously described [Ladetto et al, BBMT 2000; Brüggemann et al, Blood 2006], according to the criteria of the European Study Group on MRD detection [van der Velden et al, Leukemia 2007], at two experienced MRD laboratories (Kiel, DE, 238 samples; Torino, IT, 143 samples). NGS was performed at the Sequenta facilities in San Francisco, CA, USA. Using universal primer sets, we amplified IGH variable, diversity, and joining gene segments from genomic DNA. Amplified products were sequenced to obtain a high degree of coverage (14 reads per each IGH molecule) and analyzed using standardized algorithms for clonotype determination. Tumor-specific clonotypes were identified for each patient based on their high-frequency in DG sample and then quantitated in FU samples. A quantitative and standardized measure of clone level among all leukocytes was determined using internal reference DNA. NGS analysis was performed independently and data were blinded until comparison. Comparability of MRD results by RQ-PCR and NGS was assessed by means of bivariate correlations between methods analysis (software R 2.15.1 package irr). Discordances were classified as follows: a positive/negative discordance was defined as major when the positive result was >1 E-05 and minor when ≤1 E-05; a quantitative discordance was defined as the presence of two positive results with a quantitative discrepancy >1 log. Results. 51 pts (93%) were evaluable with at least one tool (RQ-PCR 45, NGS 49), 43 (78%) with both and 4 (7%) with none. Disease-specific success rates are shown in Tab. 1. Overall, 333 samples (87%) were evaluated with at least one tool (RQ-PCR 282, NGS 319) and 268 (70%) with both. The latter group was thus comparable in terms of MRD output. A correlation analysis between RQ-PCR and NGS results is shown in Fig. 1. Overall a significant concordance was observed (p Conclusions. NGS is a feasible tool for IGH-based MRD monitoring in ALL, MCL and MM, in selected cases reaching similar sensitivities compared to standardized RQ-PCR. Good concordance was seen in the vast majority of cases. However, disease-specific pitfalls (clonal evolution, somatic hypermutations, frequency of complete IGH rearrangements) have to be considered for both methods and prospective comparative analysis of unselected cases is required to verify the clinical impact of NGS-based MRD assessment. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.