A Palumbo
University of Turin
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by A Palumbo.
The New England Journal of Medicine | 2015
Abstr Act; Henk M. Lokhorst; Torben Plesner; Hareth Nahi; Peter Gimsing; Markus Hansson; Ulrik V. Lassen; Jakub Krejcik; A Palumbo; Tahamtan Ahmadi; I. Khan; Jianping Wang; N. Losic; Steen Lisby; Linda Basse; Nikolai C. Brun
BACKGROUND Multiple myeloma cells uniformly overexpress CD38. We studied daratumumab, a CD38-targeting, human IgG1κ monoclonal antibody, in a phase 1-2 trial involving patients with relapsed myeloma or relapsed myeloma that was refractory to two or more prior lines of therapy. METHODS In part 1, the dose-escalation phase, we administered daratumumab at doses of 0.005 to 24 mg per kilogram of body weight. In part 2, the dose-expansion phase, 30 patients received 8 mg per kilogram of daratumumab and 42 received 16 mg per kilogram, administered once weekly (8 doses), twice monthly (8 doses), and monthly for up to 24 months. End points included safety, efficacy, and pharmacokinetics. RESULTS No maximum tolerated dose was identified in part 1. In part 2, the median time since diagnosis was 5.7 years. Patients had received a median of four prior treatments; 79% of the patients had disease that was refractory to the last therapy received (64% had disease refractory to proteasome inhibitors and immunomodulatory drugs and 64% had disease refractory to bortezomib and lenalidomide), and 76% had received autologous stem-cell transplants. Infusion-related reactions in part 2 were mild (71% of patients had an event of any grade, and 1% had an event of grade 3), with no dose-dependent adverse events. The most common adverse events of grade 3 or 4 (in ≥ 5% of patients) were pneumonia and thrombocytopenia. The overall response rate was 36% in the cohort that received 16 mg per kilogram (15 patients had a partial response or better, including 2 with a complete response and 2 with a very good partial response) and 10% in the cohort that received 8 mg per kilogram (3 had a partial response). In the cohort that received 16 mg per kilogram, the median progression-free survival was 5.6 months (95% confidence interval [CI], 4.2 to 8.1), and 65% (95% CI, 28 to 86) of the patients who had a response did not have progression at 12 months. CONCLUSIONS Daratumumab monotherapy had a favorable safety profile and encouraging efficacy in patients with heavily pretreated and refractory myeloma. (Funded by Janssen Research and Development and Genmab; ClinicalTrials.gov number, NCT00574288.).
The New England Journal of Medicine | 2016
Philippe Moreau; Tamas Masszi; Norbert Grzasko; Nizar J. Bahlis; Markus Hansson; Luděk Pour; Irwindeep Sandhu; Peter Ganly; Bartrum Baker; Sharon Jackson; Anne-Marie Stoppa; David R Simpson; Peter Gimsing; A Palumbo; L. Garderet; Michele Cavo; Shaji Kumar; Cyrille Touzeau; Francis Buadi; Jacob P. Laubach; Deborah Berg; Jianchang Lin; A. Di Bacco; Ai-Min Hui; H van de Velde; Paul G. Richardson; Eric Kupperman; Allison Berger; Larry Dick; Mark Williamson
BACKGROUND Ixazomib is an oral proteasome inhibitor that is currently being studied for the treatment of multiple myeloma. METHODS In this double-blind, placebo-controlled, phase 3 trial, we randomly assigned 722 patients who had relapsed, refractory, or relapsed and refractory multiple myeloma to receive ixazomib plus lenalidomide-dexamethasone (ixazomib group) or placebo plus lenalidomide-dexamethasone (placebo group). The primary end point was progression-free survival. RESULTS Progression-free survival was significantly longer in the ixazomib group than in the placebo group at a median follow-up of 14.7 months (median progression-free survival, 20.6 months vs. 14.7 months; hazard ratio for disease progression or death in the ixazomib group, 0.74; P=0.01); a benefit with respect to progression-free survival was observed with the ixazomib regimen, as compared with the placebo regimen, in all prespecified patient subgroups, including in patients with high-risk cytogenetic abnormalities. The overall rates of response were 78% in the ixazomib group and 72% in the placebo group, and the corresponding rates of complete response plus very good partial response were 48% and 39%. The median time to response was 1.1 months in the ixazomib group and 1.9 months in the placebo group, and the corresponding median duration of response was 20.5 months and 15.0 months. At a median follow-up of approximately 23 months, the median overall survival has not been reached in either study group, and follow-up is ongoing. The rates of serious adverse events were similar in the two study groups (47% in the ixazomib group and 49% in the placebo group), as were the rates of death during the study period (4% and 6%, respectively); adverse events of at least grade 3 severity occurred in 74% and 69% of the patients, respectively. Thrombocytopenia of grade 3 and grade 4 severity occurred more frequently in the ixazomib group (12% and 7% of the patients, respectively) than in the placebo group (5% and 4% of the patients, respectively). Rash occurred more frequently in the ixazomib group than in the placebo group (36% vs. 23% of the patients), as did gastrointestinal adverse events, which were predominantly low grade. The incidence of peripheral neuropathy was 27% in the ixazomib group and 22% in the placebo group (grade 3 events occurred in 2% of the patients in each study group). Patient-reported quality of life was similar in the two study groups. CONCLUSIONS The addition of ixazomib to a regimen of lenalidomide and dexamethasone was associated with significantly longer progression-free survival; the additional toxic effects with this all-oral regimen were limited. (Funded by Millennium Pharmaceuticals; TOURMALINE-MM1 ClinicalTrials.gov number, NCT01564537.).
Leukemia | 2014
Enrique M. Ocio; Paul G. Richardson; S V Rajkumar; A Palumbo; M.V. Mateos; Robert Z. Orlowski; Shaji Kumar; Saad Z Usmani; D. Roodman; Ruben Niesvizky; Hermann Einsele; Kenneth C. Anderson; M. A. Dimopoulos; Hervé Avet-Loiseau; U-H Mellqvist; Ingemar Turesson; Giampaolo Merlini; Rik Schots; P.L. McCarthy; Leif Bergsagel; Chor Sang Chim; Juan José Lahuerta; Jatin J. Shah; A. Reiman; Joseph R. Mikhael; Sonja Zweegman; S. Lonial; Raymond L. Comenzo; Wee Joo Chng; P. Moreau
Treatment in medical oncology is gradually shifting from the use of nonspecific chemotherapeutic agents toward an era of novel targeted therapy in which drugs and their combinations target specific aspects of the biology of tumor cells. Multiple myeloma (MM) has become one of the best examples in this regard, reflected in the identification of new pathogenic mechanisms, together with the development of novel drugs that are being explored from the preclinical setting to the early phases of clinical development. We review the biological rationale for the use of the most important new agents for treating MM and summarize their clinical activity in an increasingly busy field. First, we discuss data from already approved and active agents (including second- and third-generation proteasome inhibitors (PIs), immunomodulatory agents and alkylators). Next, we focus on agents with novel mechanisms of action, such as monoclonal antibodies (MoAbs), cell cycle-specific drugs, deacetylase inhibitors, agents acting on the unfolded protein response, signaling transduction pathway inhibitors and kinase inhibitors. Among this plethora of new agents or mechanisms, some are specially promising: anti-CD38 MoAb, such as daratumumab, are the first antibodies with clinical activity as single agents in MM. Moreover, the kinesin spindle protein inhibitor Arry-520 is effective in monotherapy as well as in combination with dexamethasone in heavily pretreated patients. Immunotherapy against MM is also being explored, and probably the most attractive example of this approach is the combination of the anti-CS1 MoAb elotuzumab with lenalidomide and dexamethasone, which has produced exciting results in the relapsed/refractory setting.
Leukemia | 2014
H. Ludwig; Jesús F. San Miguel; M. A. Dimopoulos; A Palumbo; R Garcia Sanz; R Powles; Suzanne Lentzsch; W Ming Chen; Jian Hou; Artur Jurczyszyn; K Romeril; Roman Hájek; Evangelos Terpos; Kazuyuki Shimizu; Douglas E. Joshua; Varnia Hungria; A Rodriguez Morales; Dina Ben-Yehuda; Pia Sondergeld; Elena Zamagni; Brian G. M. Durie
Recent developments have led to remarkable improvements in the assessment and treatment of patients with multiple myeloma (MM). New technologies have become available to precisely evaluate the biology and extent of the disease, including information about cytogenetics and genetic abnormalities, extramedullary manifestations and minimal residual disease. New, more effective drugs have been introduced into clinical practice, which enable clinicians to significantly improve the outcome of patients but also pose new challenges for the prevention and management of their specific side effects. Given these various new options and challenges, it is important to identify the minimal requirements for diagnosis and treatment of patients, as access to the most sophisticated advances may vary depending on local circumstances. Here, we propose the minimal requirements and possible options for diagnosis, monitoring and treatment of patients with multiple myeloma.
Nature Reviews Clinical Oncology | 2006
T. Caravita; P. De Fabritiis; A Palumbo; S. Amadori; Mario Boccadoro
Proteasome inhibition represents a new anticancer approach, with the potential effect of arresting tumor growth, metastasis and angiogenesis through the activation of multiple mechanisms. Bortezomib is a biologically active agent, producing predictable, dose-related and reversible proteasome inhibition; it has shown antitumor activity in various malignancies and is the first proteasome inhibitor to be used in clinical practice. Several trials demonstrated that bortezomib is relatively well tolerated, causing manageable nonhematologic and hematologic toxicity. The drug was approved in 2003 by the FDA for the treatment of patients with multiple myeloma who had received at least two prior therapies and demonstrated disease progression on the last therapy; its application was expanded recently for second-line treatment. This article summarizes the principal clinical trials of bortezomib and discusses its efficacy in solid and hematologic tumors.
Leukemia | 2016
Jacob P. Laubach; Laurent Garderet; Anuj Mahindra; Gösta Gahrton; Jo Caers; Orhan Sezer; Peter M. Voorhees; Xavier Leleu; Hans Erik Johnsen; M. Streetly; Artur Jurczyszyn; H. Ludwig; Ulf-Henrik Mellqvist; Wee Joo Chng; Linda M. Pilarski; Hermann Einsele; Jian Hou; Ingemar Turesson; Elena Zamagni; Chor Sang Chim; Amitabha Mazumder; Jan Westin; Jin Lu; Tony Reiman; Sigurdur Y. Kristinsson; Doug Joshua; Murielle Roussel; P. O'Gorman; Evangelos Terpos; P.L. McCarthy
The prognosis for patients multiple myeloma (MM) has improved substantially over the past decade with the development of new, more effective chemotherapeutic agents and regimens that possess a high level of anti-tumor activity. In spite of this important progress, however, nearly all MM patients ultimately relapse, even those who experience a complete response to initial therapy. Management of relapsed MM thus represents a vital aspect of the overall care for patients with MM and a critical area of ongoing scientific and clinical research. This comprehensive manuscript from the International Myeloma Working Group provides detailed recommendations on management of relapsed disease, with sections dedicated to diagnostic evaluation, determinants of therapy, and general approach to patients with specific disease characteristics. In addition, the manuscript provides a summary of evidence from clinical trials that have significantly impacted the field, including those evaluating conventional dose therapies, as well as both autologous and allogeneic stem cell transplantation. Specific recommendations are offered for management of first and second relapse, relapsed and refractory disease, and both autologous and allogeneic transplant. Finally, perspective is provided regarding new agents and promising directions in management of relapsed MM.
Bone Marrow Transplantation | 1998
Ignazio Majolino; Paolo Corradini; Rosanna Scimè; Armando Santoro; Corrado Tarella; Am Cavallaro; A Palumbo; A Indovina; Daniele Caracciolo; Mario Boccadoro; R Marcenò; A Pileri
In multiple myeloma (MM), allogeneic bone marrow transplantation may produce complete and durable responses, but is accompanied by significant transplant-related mortality (TRM). To assess feasibility and possible advantages offered by the use of allogeneic, growth factor-primed PBSC instead of marrow, we analyzed the data of 10 patients with MM (IgG = 6, IgA = 1, BJ = 2, non-secreting = 1; stage II = 1, stage III = 8, plasma-cell leukemia = 1) who received an allogeneic transplant with PBSC. Their age ranged between 35 and 53 years (median 45). All were HLA-identical to their sibling donors. Prior to allograft, six patients received standard-dose chemotherapy (DAV or CY-Dexa) and four a sequential intensified scheme with autologous PBSC support. At the time of transplantation, three patients were in CR, three in PR, three had refractory disease, one progressive disease. Patients were conditioned with busulfan–melphalan (n = 9) or busulfan–cyclophosphamide (n = 1), and were allografted with unmanipulated PBSC obtained by apheresis after treatment with G-CSF alone (n = 6) or GM-CSF followed by G-CSF (n = 4). All patients engrafted, with 0.5 × 109/l PMN and 50 × 109/l platelets on (median) day 13. Four patients had ⩾grade II acute GVHD (grade II in 3, grade III in 1). Following allograft, CR was achieved in 71% patients. Eight are currently alive, with six in CR at a median of 18.5 months (range 7–28) from the transplant. Two patients died, 1 and 4 months from the allograft, respectively, and one is alive with progression. A PCR analysis of IgH rearrangement showed that residual disease was no more molecularly detectable in four out of seven evaluated patients following allograft. The results suggest that PBSC may improve the therapeutic efficacy of allogeneic transplant in MM, not only by a reduction of TRM but also by an improvement of rate and quality of response.
Leukemia | 2015
Simone Ferrero; Marco Ladetto; Daniela Drandi; Federica Cavallo; Elisa Genuardi; Marina Urbano; Simona Caltagirone; Mariella Grasso; Fausto Rossini; Tommasina Guglielmelli; C Cangialosi; Anna Marina Liberati; Vincenzo Callea; T Carovita; C Crippa; L. De Rosa; Francesco Pisani; Antonietta Falcone; Patrizia Pregno; Stefania Oliva; Carolina Terragna; Pellegrino Musto; Roberto Passera; M Boccadoro; A Palumbo
Polymerase chain reaction (PCR)-based minimal residual disease (MRD) analysis is a useful prognostic tool in multiple myeloma (MM), although its long-term impact still needs to be addressed. This report presents the updated results of the GIMEMA-VEL-03-096 trial. Thirty-nine MM patients receiving bortezomib–thalidomide–dexamethasone after autologous transplantation were monitored for MRD by both nested and real-time quantitative-PCR until relapse. Our data confirm the strong impact of MRD on survival: overall survival was 72% at 8 years median follow-up for patients in major MRD response versus 48% for those experiencing MRD persistence (P=0.041). In addition, MRD kinetics resulted predictive for relapse: indeed median remission duration was not reached for patients in major MRD response, 38 months for those experiencing MRD reappearance and 9 months for patients with MRD persistence (P<0.001). Moreover: (1) 26 patients achieving major MRD response (67%) benefit of excellent disease control (median TNT: 42 months); (2) MRD reappearance heralds relapse, with a TNT comparable to that of MRD persistence (9 versus 10 months, P=0.706); (3) the median lag between MRD reappearance and need for salvage treatment is 9 months. These results suggest the usefulness of a long-term MRD monitoring in MM patients and the need for maintenance or pre-emptive treatments ensuring durable responses.
Leukemia | 2011
Federica Cavallo; Sara Bringhen; Giuseppe Milone; D. Ben-Yehuda; Arnon Nagler; E. Calabrese; Nicola Cascavilla; Vittorio Montefusco; B. Lupo; Anna Marina Liberati; C. Crippa; Fausto Rossini; Roberto Passera; F Patriarca; Anna Maria Cafro; Paola Omedè; Angelo Michele Carella; Jacopo Peccatori; Lucio Catalano; Tommaso Caravita; Pellegrino Musto; Mt Petrucci; Mario Boccadoro; A Palumbo
Lenalidomide has raised concerns regarding its potential impact on the ability to collect stem cells for autologous stem cell transplantation, especially after prolonged exposure. The use of cyclophosphamide plus granulocyte colony-stimulating factor (G-CSF) to mobilize peripheral blood stem cells may overcome this concern. In newly diagnosed multiple myeloma (MM) patients, we investigated the influence of lenalidomide on stem cell collection. In a prospective study, 346 patients received four cycles of lenalidomide–dexamethasone (Rd). Stem cells were mobilized with cyclophosphamide and G-CSF. Patients failing to collect a minimum of 4 × 106 CD34+/kg cells received a second mobilization course. After mobilization, a median yield of 8.7 × 106 CD34+/kg was obtained from patients receiving Rd induction. After first mobilization, inadequate yield was observed in 21% of patients, whereas only 9% of patients failed to collect the target yield after the second mobilization attempt. In conclusion, we confirm that a short induction with lenalidomide allowed sufficient stem cells collection to perform autologous transplantation in 91% of newly diagnosed patients.
Journal of Thrombosis and Haemostasis | 2006
A Palumbo; Cecilia Rus; Jerome B. Zeldis; F. Rodeghiero; Mario Boccadoro
artery. J Pineal Res 2001; 31: 281–8. 12 Bertolucci C, Pinotti M, Colognesi I, Foa A, Bernardi F, Portaluppi F. Circadian rhythms in mouse blood coagulation. J Biol Rhythms 2005; 20: 219–24. 13 Iversen PO, Dahm A, Sandset PM. Circadian variations within the hemostatic and fibrinolytic systems, with emphasis on venous thromboembolism. In: Columbus F, ed. Trend in Chronobiology Research. New York: Nova Science Publishers Inc, in press.