422 results on '"Alessandra Iurlo"'
Search Results
2. Incidence of blast phase in myelofibrosis according to anemia severity
- Author
-
Barbara Mora, Margherita Maffioli, Elisa Rumi, Paola Guglielmelli, Marianna Caramella, Andrew Kuykendall, Francesca Palandri, Alessandra Iurlo, Valerio De Stefano, Jean‐Jacques Kiladjian, Elena M. Elli, Nicola Polverelli, Jason Gotlib, Francesco Albano, Richard T. Silver, Giulia Benevolo, David M. Ross, Timothy Devos, Oscar Borsani, Tiziano Barbui, Matteo G. Della Porta, Lorenza Bertù, Rami Komrokji, Alessandro M. Vannucchi, and Francesco Passamonti
- Subjects
acute myeloid leukemia ,essential thrombocythemia ,myelofibrosis ,polycythemia vera ,ruxolitinib ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Myelofibrosis (MF) is a clonal malignancy frequently characterized by anemia and in 10%–20% of cases it can evolve into blast phase (BP). Anemia in MF is associated with reduced survival and ‐in primary MF‐ also with an increased probability of BP. Conventional treatments for anemia have limited effectiveness in MF. Within a dataset of 1752 MF subjects largely unexposed to ruxolitinib (RUX), BP incidence was 2.5% patients per year (p‐y). This rate reached respectively 4.3% and 4.5% p‐y in case of patients with common terminology criteria for adverse events (CTCAE) grade 3/4 and grade 2 anemia, respectively, that represented together 32% of the cohort. Among 273 MF cases treated with RUX, BP incidence was 2.89% p‐y and it reached 4.86% p‐y in subjects who started RUX with CTCAE grade 2 anemia (one third of total). Within patients with red blood cell transfusion‐dependency at 6 months of RUX (21% of the exposed), BP rate was 4.2% p‐y. Our study highlights a relevant incidence of BP in anemic MF patients, with a similar rate whether treated with or without RUX. These findings will help treating physicians to make decisions on the safety profile of innovative anemia treatments.
- Published
- 2023
- Full Text
- View/download PDF
3. Neutrophilic and eosinophilic dermatoses associated with hematological malignancy
- Author
-
Carlo Alberto Maronese, Federica Derlino, Chiara Moltrasio, Daniele Cattaneo, Alessandra Iurlo, and Angelo Valerio Marzano
- Subjects
neutrophilic ,eosinophilic ,dermatoses ,pyoderma ,sweet ,Medicine (General) ,R5-920 - Abstract
Cutaneous manifestations of hematologic malignancy represent both a clinical challenge for the treating physician and a pathophysiological model for advancing the knowledge on individual neoplasms. Indeed, a growing body of evidence supports the concept of recurrent molecular defects associating with specific clinical features, as best exemplified by VEXAS. Herein neutrophilic and eosinophilic dermatoses of potential interest for both hematologists and dermatologists will be reviewed, including subcorneal pustular dermatosis-type IgA pemphigus, neutrophilic eccrine hidradenitis, Sweet’s syndrome as well as myelodysplasia cutis and VEXAS, pyoderma gangrenosum, eosinophilic annular erythema, eosinophilic dermatosis of hematological malignancy, Wells syndrome and cutaneous involvement in hypereosinophilic syndromes. Possible management approaches are discussed for each, emphasizing scenarios that require treatment of the underlying condition to achieve remission at the skin level.
- Published
- 2024
- Full Text
- View/download PDF
4. Determinants of Covid19 disease and of survival after Covid19 in MPN patients treated with ruxolitinib
- Author
-
Francesca Palandri, Elena M. Elli, Giuseppe Auteri, Massimiliano Bonifacio, Giulia Benevolo, Florian H. Heidel, Simona Paglia, Malgorzata M. Trawinska, Costanza Bosi, Elena Rossi, Mario Tiribelli, Alessia Tieghi, Alessandra Iurlo, Nicola Polverelli, Giovanni Caocci, Gianni Binotto, Francesco Cavazzini, Eloise Beggiato, Daniela Cilloni, Caterina Tatarelli, Francesco Mendicino, Maurizio Miglino, Monica Bocchia, Monica Crugnola, Camilla Mazzoni, Andrea D. Romagnoli, Giovanni Rindone, Sara Ceglie, Alessandra D’Addio, Eleonora Santoni, Daniele Cattaneo, Daniela Bartoletti, Roberto M. Lemoli, Mauro Krampera, Antonio Cuneo, Gianpietro C. Semenzato, Roberto Latagliata, Elisabetta Abruzzese, Nicola Vianelli, Michele Cavo, Alessandro Andriani, Valerio De Stefano, Giuseppe A. Palumbo, and Massimo Breccia
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
5. A New Algorithm Integrating Molecular Response, Toxicity, and Plasma Level Measures for Ponatinib Dose Choice in Patients Affected by Chronic Myeloid Leukemia
- Author
-
Sara Galimberti, Elisabetta Abruzzese, Giacomo Luci, Claudia Baratè, Luigia Luciano, Alessandra Iurlo, Giovanni Caocci, Riccardo Morganti, Fabio Stefanelli, and Antonello Di Paolo
- Subjects
chronic myeloid leukemia ,ponatinib ,therapeutic drug monitoring ,molecular response ,toxicity ,Pharmacy and materia medica ,RS1-441 - Abstract
Ponatinib may be effective in chronic myeloid leukemia (CML) patients after failure of first/second line therapies. Although its efficacy for minimum plasma concentrations (Cmin) is >21.3 ng/mL (equal to 40 nM), ponatinib may cause adverse events (AE) that require dose optimization. The present study was aimed at investigating any possible correlations among ponatinib dose, plasma concentration, molecular response (MR), and tolerability in a real-world setting. Clinical and laboratory records (including MR and drug plasma concentrations) of 32 CML patients treated with ponatinib were harvested and analyzed. Twenty-seven patients (71%) had ponatinib Cmin values > 21.3 ng/mL, but Cmin values > 10.7 ng/mL (considered efficacious in BCR-Abl unmutated patients) were achieved by 80% of the patients receiving ≥30 mg/day and 45% of the subjects treated with 15 mg/day. No significant correlations were identified among clinical efficacy, tolerability, daily dose, and plasma concentration. Notably, patients who underwent dose tapering for tolerability or safety reasons did not experience treatment failure. In a real-world setting, adjustment of ponatinib daily doses lower than those registered may maintain therapeutic efficacy while reducing the risk of vascular events and improving tolerability. Further studies are warranted to confirm the present results in a larger cohort of patients.
- Published
- 2024
- Full Text
- View/download PDF
6. Impact on mental health, disease management, and socioeconomic modifications in hematological patients during the COVID-19 pandemic in Italy
- Author
-
Marianna De Muro, Annelot Julia Janssen, Sergio Amadori, Paolo de Fabritiis, Dante Sabatino, Pasquale Niscola, Lorenza Torti, Malgorzata Monika Trawinska, Cristiano Tesei, Felice Bombaci, Mario Tarricone, Monica Bocchia, Carmen Fava, Sara Galimberti, Alessandra Iurlo, Luigia Luciano, and Elisabetta Abruzzese
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background: Hematological patients are a highly vulnerable population with an increased risk of developing severe COVID-19 symptoms due to their immunocompromised status. COVID-19 has proven to cause serious mental health issues, such as stress, anxiety, and depression in the general population. However, data on the psycho-social impact of COVID-19 on hematological patients are lacking. Objectives: This study aims to examine the psychological well-being of hematological patients in Italy during the initial period of the COVID-19 pandemic. Furthermore, it seeks to explore the association between modifications in the management of hematological diseases and employment status of these patients during the COVID-19 pandemic and the resulting mental health outcomes. Design and Methods: A survey using the DASS-21 questionnaire was administered to 1105 hematological patients. Data analysis was conducted using the R software, and logistic regression analysis was performed to predict the association between hematological patient/general population and employment status with DASS scores. Results: The hematological patient population reported significantly higher levels of depression (OR 0.947, 95% CI 0.966–0.982, p
- Published
- 2023
- Full Text
- View/download PDF
7. Breakthrough infections in MPN-COVID vaccinated patients
- Author
-
Tiziano Barbui, Alessandra Carobbio, Arianna Ghirardi, Alessandra Iurlo, Valerio De Stefano, Marta Anna Sobas, Elisa Rumi, Elena Maria Elli, Francesca Lunghi, Mercedes Gasior Kabat, Beatriz Cuevas, Paola Guglielmelli, Massimiliano Bonifacio, Monia Marchetti, Alberto Alvarez-Larran, Laura Fox, Marta Bellini, Rosa Daffini, Giulia Benevolo, Gonzalo Carreno-Tarragona, Andrea Patriarca, Haifa Kathrin Al-Ali, Maria Marcio Miguel Andrade-Campos, Francesca Palandri, Claire Harrison, Maria Angeles Foncillas, Santiago Osorio, Steffen Koschmieder, Elena Magro Mazo, Jean-Jacques Kiladjian, Estefanía Bolaños Calderón, Florian H. Heidel, Keina Quiroz Cervantes, Martin Griesshammer, Valentin Garcia-Gutierrez, Alberto Marin Sanchez, Juan Carlos Hernandez-Boluda, Emma Lopez Abadia, Giuseppe Carli, Miguel Sagues Serrano, Rajko Kusec, Blanca Xicoy Cirici, Margarita Guenova, Begona Navas Elorza, Anna Angona, Edyta Cichocka, Anna Kulikowska de Nałęcz, Daniele Cattaneo, Cristina Bucelli, Silvia Betti, Oscar Borsani, Fabrizio Cavalca, Sara Carbonell, Natalia Curto-Garcia, Lina Benajiba, Alessandro Rambaldi, and Alessandro Maria Vannucchi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2022
- Full Text
- View/download PDF
8. P663: MULTICENTER, PROSPECTIVE AND RETROSPECTIVE OBSERVATIONAL COHORT STUDY OF PONATINIB IN PATIENTS WITH CML IN ITALY: LONG-TERM FOLLOW-UP RESULTS OF THE OITI TRIAL
- Author
-
Massimo Breccia, Alessandra Iurlo, Felicetto Ferrara, Immacolata Attolico, Alessandra Malato, Massimiliano Bonifacio, Anna Rita Scortechini, Elisabetta Abruzzese, Grazia Sanpaolo, Nicola DI Renzo, Monia Lunghi, Stefana Impera, Fausto Castagnetti, Mario Tiribelli, Marco Santoro, Francesca Lunghi, Alessandro Maggi, Valeria Sargentini, Alfonso Piciocchi, Claudia Tringali, and Luigiana Luciano
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
9. P667: RISK OF PROGRESSION IN CHRONIC PHASE-CHRONIC MYELOID LEUKEMIA PATIENTS ELIGIBLE FOR TYROSINE KINASE INHIBITOR DISCONTINUATION: FINAL ANALYSIS OF THE TFR-PRO STUDY
- Author
-
Giovanni Paolo Maria Zambrotta, Sarit Assouline, Franck E. Nicolini, Elisabetta Abruzzese, Ester Pungolino, Maria Cristina Miggiano, Chiara Elena, Alberto Alvarez Larran, Ana Triguero Moreno, Alessandra Iurlo, Cristina Bucelli, Francesca Lunghi, Isabella Capodanno, Margherita Maffioli, Sara Galimberti, Giovanni Caocci, Fabio Stagno, Susanne Saussele, Carmen Fava, Philipp Le Coutre, Massimiliano Bonifacio, Veronica Guglielmana, Federica Colombo, Laura Antolinni, and Carlo Gambacorti-Passerini
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
10. P686: REAL-WORLD EFFICACY PROFILE OF ASCIMINIB IN AN ITALIAN, MULTI-RESISTANT CHRONIC-PHASE CHRONIC MYELOID LEUKEMIA (CP-CML) PATIENT POPULATION
- Author
-
Massimo Breccia, Antonella Russo Rossi, Valentina Giai, Bruno Martino, Carmen Fava, Mario Annunziata, Elisabetta Abruzzese, Gianni Binotto, Claudia Baratè, Aurelio Pio Nardozza, Alessandra Misto, Paola Coco, Valeria Calafiore, Maria Cristina Carraro, Federica Cattina, Francesco Cavazzini, Maria Teresa Corsetti, Lara Crucitti, Monica Crugnola, Paolo Ditonno, Ambra DI Veroli, Anna Ermacora, Felicetto Ferrara, Angelo Genua, Antonella Gozzini, Stefana Impera, Alessandra Iurlo, Luciano Levato, Luigiana Luciano, Maria Cristina Miggiano, Marco De Gobbi, Marco Santoro, Barbara Scappini, Anna Rita Scortechini, Andrea Patriarca, Serena Rosati, Sabina Russo, Rosaria Sancetta, Grazia Sanpaolo, Teresa Maria Santeramo, Silvia Sibilla, Federica Sorà, Paolo Sportoletti, Fabio Stagno, Elena Trabacchi, and Fausto Castagnetti
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
11. P989: NEWLY IDENTIFIED ROLES FOR PIEZO1 MECHANOSENSOR IN CONTROLLING NORMAL MEGAKARYOCYTE DEVELOPMENT AND IN PRIMARY MYELOFIBROSIS
- Author
-
Vittorio Abbonante, Iris A. Karkempetzaki, Christian A. DI Buduo, Nasi Huang, Daniele Cattaneo, Christina Ward, Shinobu Matsuura, Alessandra Iurlo, Katya Ravid, and Alessandra Balduini
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
12. P1041: LEUKOCYTOSIS SELECTS A SUBGROUP OF LOW-RISK PV PATIENTS IN WHOM PHLEBOTOMY-ALONE MAY BE INSUFFICIENT
- Author
-
Alessandra Carobbio, Alessandro Vannucchi, Valerio De Stefano, Arianna Ghirardi, Greta Carioli, Arianna Masciulli, Elena Rossi, Fabio Ciceri, Massimiliano Bonifacio, Alessandra Iurlo, Francesca Palandri, Giulia Benevolo, Fabrizio Pane, Alessandra Ricco, Giuseppe Carli, Marianna Caramella, Davide Rapezzi, Caterina Musolino, Sergio Siragusa, Elisa Rumi, Andrea Patriarca, Nicola Cascavilla, Barbara Mora, Emma Cacciola, Carmela Mannarelli, Giuseppe Gaetano Loscocco, Paola Guglielmelli, Francesca Gesullo, Silvia Betti, Francesca Lunghi, Luigi Scaffidi, Cristina Bucelli, Nicola Vianelli, Marta Bellini, Maria Chiara Finazzi, Gianni Tognoni, Alessandro Rambaldi, and Tiziano Barbui
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
13. P1055: BROMODOMAIN AND EXTRA-TERMINAL (BET) INHIBITOR INCB057643 (LIMBER-103) IN PATIENTS (PTS) WITH RELAPSED OR REFRACTORY MYELOFIBROSIS (R/R MF) AND OTHER ADVANCED MYELOID NEOPLASMS: A PHASE 1 STUDY
- Author
-
Justin M. Watts, Anthony M. Hunter, Alessandra Iurlo, Blanca Xicoy, Francesca Palandri, Brandi Reeves, Alessandro Vannucchi, Prithviraj Bose, Rosa Ayala Diaz, Anna B. Halpern, Xuejun Chen, Francis Seguy, Feng Zhou, Fred Zheng, and Pankit Vachhani
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
14. P1002: PROTEOMIC SCREENING IDENTIFIES MEGAKARYOCYTE DERIVED PF4/CXCL4 AS A CRITICAL DRIVER OF MYELOFIBROSIS
- Author
-
Alessandro Malara, Vittorio Abbonante, Francesca Rossella Calledda, Daniele Capitanio, Daniele Cattaneo, Alessandra Iurlo, Cecilia Gelfi, and Alessandra Balduini
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
15. P1028: MYELOFIBROSIS (MF) TREATED WITH RUXOLITINIB (RUX) MONOTHERAPY: PREDICTORS OF EARLY DISCONTINUATION AND DEATH ON TREATMENT. THE SHORT-TERM RUX PROGNOSTIC MODEL (STR-PM)
- Author
-
Francesca Palandri, Giuseppe Auteri, Alessandra Iurlo, Elena Maria Elli, Simona Paglia, Massimiliano Bonifacio, Mario Tiribelli, Malgorzata Monica Trawinska, Nicola Polverelli, Giulia Benevolo, Alessia Tieghi, Florian Heidel, Fabrizio Cavalca, Giovanni Caocci, Eloise Beggiato, Gianni Binotto, Francesco Cavazzini, Maurizio Miglino, Costanza Bosi, Monica Crugnola, Monica Bocchia, Bruno Martino, Novella Pugliese, Marta Venturi, Camilla Mazzoni, Alessandro Isidori, Daniele Cattaneo, Mauro Krampera, Fabrizio Pane, Daniela Cilloni, Gianpietro Semenzato, Roberto M. Lemoni, Antonio Cuneo, Elisabetta Abruzzese, Nicola Vianelli, Michele Cavo, Giuseppe Palumbo, and Massimo Breccia
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
16. Management of Myelofibrosis during Treatment with Ruxolitinib: A Real-World Perspective in Case of Resistance and/or Intolerance
- Author
-
Massimo Breccia, Francesca Palandri, Paola Guglielmelli, Giuseppe Alberto Palumbo, Alessandra Malato, Francesco Mendicino, Alessandra Ricco, Emanuela Sant’Antonio, Mario Tiribelli, and Alessandra Iurlo
- Subjects
anemia ,infection ,intolerance ,myelofibrosis ,non-melanoma skin cancer ,resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The development and approval of ruxolitinib, the first JAK1/2 inhibitor indicated to treat myelofibrosis, has improved patient outcomes, with higher spleen and symptoms responses, improved quality of life, and overall survival. Despite this, several unmet needs remain, including the absence of resistance criteria, suboptimal response, the timing of allogeneic transplant, and the management of patients in case of intolerance. Here, we report the results of the second survey led by the “MPN Lab” collaboration, which aimed to report physicians’ perspectives on these topics. As in our first survey, physicians were selected throughout Italy, and we included those with extensive experience in treating myeloproliferative neoplasms and those with less experience representing clinical practice in the real world. The results presented here, summarized using descriptive analyses, highlight the need for a clear definition of response to ruxolitinib as well as recommendations to guide the management of ruxolitinib under specific conditions including anemia, thrombocytopenia, infections, and non-melanoma skin cancers.
- Published
- 2022
- Full Text
- View/download PDF
17. Dihydroorotate dehydrogenase inhibition reveals metabolic vulnerability in chronic myeloid leukemia
- Author
-
Mohammad Houshmand, Nicoletta Vitale, Francesca Orso, Alessandro Cignetti, Ivan Molineris, Valentina Gaidano, Stefano Sainas, Marta Giorgis, Donatella Boschi, Carmen Fava, Alice Passoni, Marta Gai, Massimo Geuna, Federica Sora, Alessandra Iurlo, Elisabetta Abruzzese, Massimo Breccia, Olga Mulas, Giovanni Caocci, Fausto Castagnetti, Daniela Taverna, Salvatore Oliviero, Fabrizio Pane, Marco Lucio Lolli, Paola Circosta, and Giuseppe Saglio
- Subjects
Cytology ,QH573-671 - Abstract
Abstract The development of different generations of BCR-ABL1 tyrosine kinase inhibitors (TKIs) has led to the high overall survival of chronic myeloid leukemia (CML) patients. However, there are CML patients who show resistance to TKI therapy and are prone to progress to more advanced phases of the disease. So, implementing an alternative approach for targeting TKIs insensitive cells would be of the essence. Dihydroorotate dehydrogenase (DHODH) is an enzyme in the de novo pyrimidine biosynthesis pathway that is located in the inner membrane of mitochondria. Here, we found that CML cells are vulnerable to DHODH inhibition mediated by Meds433, a new and potent DHODH inhibitor recently developed by our group. Meds433 significantly activates the apoptotic pathway and leads to the reduction of amino acids and induction of huge metabolic stress in CML CD34+ cells. Altogether, our study shows that DHODH inhibition is a promising approach for targeting CML stem/progenitor cells and may help more patients discontinue the therapy.
- Published
- 2022
- Full Text
- View/download PDF
18. Prospective monitoring of chronic myeloid leukemia patients from the time of TKI discontinuation: the fate of peripheral blood CD26+ leukemia stem cells
- Author
-
Paola Pacelli, Adele Santoni, Anna Sicuranza, Elisabetta Abruzzese, Valentina Giai, Monica Crugnola, Mario Annunziata, Sara Galimberti, Alessandra Iurlo, Luigiana Luciano, Federica Sorà, Carmen Fava, Elena Bestoso, Cristina Marzano, Alessandra Cartocci, Marzia Defina, Vincenzo Sammartano, Emanuele Cencini, Donatella Raspadori, and Monica Bocchia
- Subjects
chronic myelogenous leukemia ,treatment free remission ,CD26+ ,leukemia stem cell ,TKI ,flow cytometry ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Introduction: In chronic myeloid leukemia (CML), about half of the patients achieving a deep and stable molecular response with tyrosine kinase inhibitors (TKIs) may discontinue TKI treatment without disease recurrence. As such, treatment-free remission (TFR) has become an ambitious goal of treatment. Given the evidence that deepness and duration of molecular response are necessary but not sufficient requisites for a successful TFR, additional biological criteria are needed to identify CML patients suitable for efficacious discontinuation. Leukemia stem cells (LSCs) are supposed to be the reservoir of the disease. Previously, we demonstrated that residual circulating CD34+/CD38-/CD26+ LSCs were still detectable in a consistent number of CML patients during TFR.Methods: CML LSCs could be easily identified by flow-cytometry as they express the CD34+/CD38-/CD26+ phenotype. In this study, we explored the role of these cells and their correlation with molecular response in a cohort of 109 consecutive chronic phase CML patients prospectively monitored from the time of TKI discontinuation.Results: After a median observation time of 33 months from TKI discontinuation, 38/109 (35%) patients failed TFR after a median time of 4 months, while 71/109 (65%) patients are still in TFR. At TKI discontinuation, peripheral blood CD26+LSCs were undetectable in 48/109 (44%) patients and detectable in 61/109 (56%). No statistically significant correlation between detectable/undetectable CD26+LSCs and the rate of TFR loss was found (p = 0.616). The incidence of TFR loss based on the type of TKI treatment was statistically significant for imatinib treatment compared to that of nilotinib (p = 0.039). Exploring the behavior of CD26+LSCs during TFR, we observed fluctuating values that were very variable between patients, and they were not predictive of TFR loss.Discussion: Up to date, our results confirm that CD26+LSCs are detectable at the time of TKI discontinuation and during TFR. Moreover, at least for the observation median time of the study, the persistence of “fluctuating” values of residual CD26+LSCs does not hamper the possibility to maintain a stable TFR. On the contrary, even patients discontinuing TKI with undetectable CD26+LSCs could undergo TFR loss. Our results suggest that factors other than residual LSCs “burden” playing an active role in controlling disease recurrence. Additional studies evaluating CD26+LSCs’ ability to modulate the immune system and their interaction in CML patients with very long stable TFR are ongoing.
- Published
- 2023
- Full Text
- View/download PDF
19. Case report: Pleural effusion during tyrosine-kinase inhibitor treatment in chronic myeloid leukemia: Not only a dasatinib-related adverse event
- Author
-
Raffaella Pasquale, Cristina Bucelli, Valentina Bellani, Manuela Zappa, Alessandra Iurlo, and Daniele Cattaneo
- Subjects
adverse event ,chronic myeloid leukemia ,nilotinib ,pleural effusion ,tyrosine-kinase inhibitor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The spectrum of TKI-related adverse events (AEs) is variable. Pleural effusion (PE) is a frequent AE attributable to dasatinib treatment, while it is only rarely associated with nilotinib. The pathogenetic mechanism leading to PE during nilotinib therapy is still unknown and its management has not yet been defined. To the best of our knowledge, only a limited number of similar case reports have already been reported in the literature so far. Here, we describe the case of a 41-year-old CML patient who developed PE during first-line nilotinib, successfully treated with steroids and nilotinib permanent discontinuation. We highlight the differences among our patient and the others, proposing therapeutic strategies to solve this rare but still possible AE, of which physicians should be aware.
- Published
- 2022
- Full Text
- View/download PDF
20. Long-term follow-up of recovered MPN patients with COVID-19
- Author
-
Tiziano Barbui, Alessandra Iurlo, Arianna Masciulli, Alessandra Carobbio, Arianna Ghirardi, Giuseppe Rossi, Claire Harrison, Alberto Alvarez-Larran, Elena Maria Elli, Jean-Jaques Kiladjian, Mercedes Gasior Kabat, Alberto Marin Sanchez, Francesca Palandri, Marcio Miguel Andrade-Campos, Alessandro Maria Vannucchi, Gonzalo Carreno-Tarragona, Petros Papadopoulos, Keina Quiroz Cervantes, Maria Angeles Foncillas, Maria Laura Fox, Miguel Sagues Serrano, Elisa Rumi, Santiago Osorio, Giulia Benevolo, Andrea Patriarca, Begona Navas Elorza, Valentin Garcia-Gutierrez, Elena Magro Mazo, Francesca Lunghi, Massimiliano Bonifacio, Valerio De Stefano, Juan Carlos Hernandez-Boluda, Emma Lopez Abadia, Anna Angona, Blanca Xicoy Cirici, Marco Ruggeri, Steffen Koschmieder, Marta Anna Sobas, Beatriz Cuevas, Daniele Cattaneo, Rosa Daffini, Marta Bellini, Natalia Curto-Garcia, Marta Garrote, Fabrizio Cavalca, Lina Benajiba, Beatriz Bellosillo, Paola Guglielmelli, Oscar Borsani, Silvia Betti, Silvia Salmoiraghi, and Alessandro Rambaldi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
21. Long-term safety and efficacy of givinostat in polycythemia vera: 4-year mean follow up of three phase 1/2 studies and a compassionate use program
- Author
-
Alessandro Rambaldi, Alessandra Iurlo, Alessandro M. Vannucchi, Bruno Martino, Attilio Guarini, Marco Ruggeri, Nikolas von Bubnoff, Marianna De Muro, Mary Frances McMullin, Stefania Luciani, Vincenzo Martinelli, Axel Nogai, Vittorio Rosti, Alessandra Ricco, Paolo Bettica, Sara Manzoni, and Silvia Di Tollo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Polycythemia vera (PV) is a BCR-ABL1-negative myeloproliferative neoplasm (MPN) characterized by excessive proliferation of erythroid, myeloid, and megakaryocytic components in the bone marrow, mainly due to a Janus kinase 2 gene mutation (JAK2 V617F). Givinostat, a histone-deacetylase inhibitor that selectively targets JAK2 V617F cell growth, has demonstrated good efficacy and safety in three phase 1/2 studies in patients with PV. This manuscript focuses on the 4-year mean (2.8 year median) follow-up of an open-label, long-term study that enrolled 51 patients with PV (out of a total of 54 with MPN) who received clinical benefit from givinostat in these previous studies or on compassionate use, and who continued to receive givinostat at the last effective and tolerated dose. The primary objectives are to determine givinostat’s long-term safety and tolerability, and efficacy evaluated by the investigators according to internationally recognized response criteria. During follow-up, only 10% of PV patients reported Grade 3 treatment-related adverse events (AEs), while none had Grade 4 or 5 treatment-related AEs. The overall response rate for the duration of follow-up was always greater than 80% in patients with PV. In conclusion, givinostat demonstrated a good safety and efficacy profile in patients with PV, data supporting long-term use in this population.
- Published
- 2021
- Full Text
- View/download PDF
22. Molecular response and quality of life in chronic myeloid leukemia patients treated with intermittent TKIs: First interim analysis of OPTkIMA study
- Author
-
Michele Malagola, Alessandra Iurlo, Elisabetta Abruzzese, Massimiliano Bonifacio, Fabio Stagno, Gianni Binotto, Mariella D’Adda, Monia Lunghi, Monica Crugnola, Maria Luisa Ferrari, Francesca Lunghi, Fausto Castagnetti, Gianantonio Rosti, Roberto M. Lemoli, Rosaria Sancetta, Maria Rosaria Coppi, Maria Teresa Corsetti, Giovanna Rege Cambrin, Atelda Romano, Mario Tiribelli, Antonella Russo Rossi, Sabina Russo, Lara Aprile, Monica Bocchia, Lisa Gandolfi, Mirko Farina, Simona Bernardi, Nicola Polverelli, Aldo M. Roccaro, Antonio De Vivo, Michele Baccarani, and Domenico Russo
- Subjects
chronic myeloid leukaemia ,intermittent ,quality of life ,tyrosine kinase inhibitor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Intermittent treatment with TKIs is an option for the great majority (70%–80%) of CML patients who do not achieve a stable deep molecular response and are not eligible for treatment discontinuation. For these patients, the only alternative is to assume TKI continuously, lifelong. Methods The Italian phase III multicentric randomized OPTkIMA study started in 2015, with the aim to evaluate if a progressive de‐escalation of TKIs (imatinib, nilotinib, and dasatinib) is able to maintain the molecular response (MR3.0) and to improve Health Related Quality of Life (HRQoL). Results Up to December 2018, 166/185 (90%) elderly CML patients in stable MR3.0/MR4.0 completed the first year of any TKI intermittent schedule 1 month ON and 1 month OFF. The first year probability of maintaining the MR3.0 was 81% and 23.5% of the patients who lost the molecular response regained the MR3.0 after resuming TKI continuously. Patients’ HRQoL at baseline was better than that of matched peers from healthy population. Women was the only factor independently associated with worse baseline HRQoL (p > 0.0001). Overall, global HRQoL worsened at 6 (p
- Published
- 2021
- Full Text
- View/download PDF
23. Impact of diagnosis and treatment on response to COVID-19 vaccine in patients with BCR-ABL1-negative myeloproliferative neoplasms. A single-center experience
- Author
-
Daniele Cattaneo, Cristina Bucelli, Francesca Cavallaro, Dario Consonni, and Alessandra Iurlo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
24. Among classic myeloproliferative neoplasms, essential thrombocythemia is associated with the greatest risk of venous thromboembolism during COVID-19
- Author
-
Tiziano Barbui, Valerio De Stefano, Alberto Alvarez-Larran, Alessandra Iurlo, Arianna Masciulli, Alessandra Carobbio, Arianna Ghirardi, Alberto Ferrari, Valeria Cancelli, Elena Maria Elli, Marcio Miguel Andrade-Campos, Mercedes Gasior Kabat, Jean-Jaques Kiladjian, Francesca Palandri, Giulia Benevolo, Valentin Garcia-Gutierrez, Maria Laura Fox, Maria Angeles Foncillas, Carmen Montoya Morcillo, Elisa Rumi, Santiago Osorio, Petros Papadopoulos, Massimiliano Bonifacio, Keina Susana Quiroz Cervantes, Miguel Sagues Serrano, Gonzalo Carreno-Tarragona, Marta Anna Sobas, Francesca Lunghi, Andrea Patriarca, Begoña Navas Elorza, Anna Angona, Elena Magro Mazo, Steffen Koschmieder, Giuseppe Carli, Beatriz Cuevas, Juan Carlos Hernandez-Boluda, Emma Lopez Abadia, Blanca Xicoy Cirici, Paola Guglielmelli, Marta Garrote, Daniele Cattaneo, Rosa Daffini, Fabrizio Cavalca, Beatriz Bellosillo, Lina Benajiba, Natalia Curto-Garcia, Marta Bellini, Silvia Betti, Claire Harrison, Alessandro Rambaldi, and Alessandro Maria Vannucchi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract In a multicenter European retrospective study including 162 patients with COVID-19 occurring in essential thrombocythemia (ET, n = 48), polycythemia vera (PV, n = 42), myelofibrosis (MF, n = 56), and prefibrotic myelofibrosis (pre-PMF, n = 16), 15 major thromboses (3 arterial and 12 venous) were registered in 14 patients, of whom all, but one, were receiving LMW-heparin prophylaxis. After adjustment for the competing risk of death, the cumulative incidence of arterial and venous thromboembolic events (VTE) reached 8.5% after 60 days follow-up. Of note, 8 of 12 VTE were seen in ET. Interestingly, at COVID-19 diagnosis, MPN patients had significantly lower platelet count (p
- Published
- 2021
- Full Text
- View/download PDF
25. Ruxolitinib discontinuation syndrome: incidence, risk factors, and management in 251 patients with myelofibrosis
- Author
-
Francesca Palandri, Giuseppe Alberto Palumbo, Elena Maria Elli, Nicola Polverelli, Giulia Benevolo, Bruno Martino, Elisabetta Abruzzese, Mario Tiribelli, Alessia Tieghi, Roberto Latagliata, Francesco Cavazzini, Micaela Bergamaschi, Gianni Binotto, Monica Crugnola, Alessandro Isidori, Giovanni Caocci, Florian Heidel, Novella Pugliese, Costanza Bosi, Daniela Bartoletti, Giuseppe Auteri, Daniele Cattaneo, Luigi Scaffidi, Malgorzata Monica Trawinska, Rossella Stella, Fiorella Ciantia, Fabrizio Pane, Antonio Cuneo, Mauro Krampera, Gianpietro Semenzato, Roberto Massimo Lemoli, Alessandra Iurlo, Nicola Vianelli, Michele Cavo, Massimo Breccia, and Massimiliano Bonifacio
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
26. Treatment-Free Remission in Chronic Myeloid Leukemia Patients Treated With Low-Dose TKIs: A Feasible Option Also in the Real-Life. A Campus CML Study
- Author
-
Alessandra Iurlo, Daniele Cattaneo, Silvia Artuso, Dario Consonni, Elisabetta Abruzzese, Gianni Binotto, Monica Bocchia, Massimiliano Bonifacio, Fausto Castagnetti, Sara Galimberti, Antonella Gozzini, Miriam Iezza, Roberto Latagliata, Luigiana Luciano, Alessandro Maggi, Maria Cristina Miggiano, Patrizia Pregno, Giovanna Rege-Cambrin, Sabina Russo, Anna Rita Scortechini, Agostino Tafuri, Mario Tiribelli, Carmen Fava, Gianantonio Rosti, Robin Foa, Massimo Breccia, and Giuseppe Saglio
- Subjects
chronic myeloid leukemia ,tyrosine kinase inhibitors (TKI) ,treatment-free remission (TFR) ,low dose ,adverse event (AE) ,real life ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Treatment-free remission (TFR) has become a primary therapeutic goal in CML and is also considered feasible by international guidelines. TKIs dose reduction is often used in real-life practice to reduce adverse events, although its impact on TFR is still a matter of debate. This study aimed to explore the attitude of Italian hematologists towards prescribing TKIs at reduced doses and its impact on TFR. In September 2020, a questionnaire was sent to 54 hematology centers in Italy participating to the Campus CML network. For each patient, data on the main disease characteristics were collected. Most of the hematologists involved (64.4%) believed that low-dose TKIs should not influence TFR. Indeed, this approach was offered to 194 patients. At the time of TFR, all but 3 patients had already achieved a DMR, with a median duration of 61.0 months. After a median follow-up of 29.2 months, 138 (71.1%) patients were still in TFR. Interestingly, TFR outcome was not impaired by any of the variables examined, including sex, risk scores, BCR-ABL1 transcript types, previous interferon, type and number of TKIs used before treatment cessation, degree of DMR or median duration of TKIs therapy. On the contrary, TFR was significantly better after dose reduction due to AEs; furthermore, patients with a longer DMR duration showed a trend towards prolonged TFR. This survey indicates that low-dose TKI treatment is an important reality. While one third of Italian hematologists still had some uncertainties on TFR feasibility after using reduced doses of TKIs outside of clinical trials, TFR has often been considered a safe option even in patients treated with low-dose TKIs in the real-life setting. It should be noted that only 28.9% of our cases had a molecular recurrence, less than reported during standard dose treatment. Consequently, TFR is not impaired using low-dose TKIs.
- Published
- 2022
- Full Text
- View/download PDF
27. How many chronic myeloid leukemia patients who started a frontline second‐generation tyrosine kinase inhibitor have to switch to a second‐line treatment? A retrospective analysis from the monitoring registries of the italian medicines agency (AIFA)
- Author
-
Massimo Breccia, Pier Paolo Olimpieri, Odoardo Olimpieri, Fabrizio Pane, Alessandra Iurlo, Paolo Foggi, Alessia Cirilli, Antonietta Colatrella, Marcello Cuomo, Lucia Gozzo, Valentina Summa, Paolo Corradini, Pierluigi Russo, and the AIFA’s Monitoring Registries Group
- Subjects
chronic myeloid leukemia ,failure ,intolerance ,second‐generation TKIs ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
ABSTRACT The frequency of patients who switch to a second‐line therapy from a frontline second‐generation (2gen) tyrosine kinase inhibitor (TKI) such as dasatinib and nilotinib, is still substantially unknown. We retrospectively investigated a large series of chronic phase chronic myeloid leukemia (CP‐CML) patients initially treated with 2gen TKIs monitored through the Italian Medicines Agency (AIFA Agenzia Italiana del farmaco) registries. Overall, 2420 patients were analyzed over a period of 6 years. One hundred and fifty‐seven patients (16.3%) treated with dasatinib and 164 treated with nilotinib (11.3%) have switched to another drug, with an overall frequency of 13.2%. In the dasatinib cohort, 39.4% of patients changed treatment for failure and 36.3% for intolerance as compared to 45.7% and 27.4% respectively in the nilotinib cohort. Overall, the median time to switch due to resistance was 293 days, whereas it was 317 days in case of intolerance. Resistance was observed mainly in younger male patients with high‐risk features, while intolerance was not related to any baseline parameter. After resistance/intolerance to nilotinib, the majority of patients switched to dasatinib (53.8%) whereas in case of frontline dasatinib to ponatinib (43.2%). To the best of our knowledge these data provide the first report on the frequency of discontinuation of frontline 2gen TKIs and on the main causes and pattern of choice to a second‐line therapy in the real‐life setting.
- Published
- 2020
- Full Text
- View/download PDF
28. Pro-Inflammatory and Pro-Oxidative Changes During Nilotinib Treatment in CML Patients: Results of a Prospective Multicenter Front-Line TKIs Study (KIARO Study)
- Author
-
Anna Sicuranza, Ilaria Ferrigno, Elisabetta Abruzzese, Alessandra Iurlo, Sara Galimberti, Antonella Gozzini, Luigiana Luciano, Fabio Stagno, Antonella Russo Rossi, Nicola Sgherza, Daniele Cattaneo, Corrado Zuanelli Brambilla, Cristina Marzano, Carmen Fava, Olga Mulas, Emanuele Cencini, Adele Santoni, Vincenzo Sammartano, Alessandro Gozzetti, Luca Puccetti, and Monica Bocchia
- Subjects
CML ,TKI ,cytokines ,AOEs ,cardiovascular risk (CV risk) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Tyrosine kinase inhibitors (TKI) may offer a normal life expectancy to Chronic Myeloid Leukemia (CML) patients. However, a higher than expected incidence of arterial occlusive events (AOEs) was observed during treatment with nilotinib. We previously showed an “inflammatory status” during nilotinib that may explain the increased incidence of AOEs. Thus, we conducted this prospective KIARO study involving 186 CML patients (89 imatinib, 59 nilotinib, 38 dasatinib). Interleukin 6 (IL6), interleukin 10 (IL10), Tumor Necrosis Factor-α (TNFα), oxLDL, and high-sensitivity C-reactive protein (hs-CRP) plasma levels were measured at diagnosis and during treatment, with the aim to investigate changes in the inflammatory status favoring AOEs of each patient. Clinical and biochemical pro-atherothrombotic profiles and the 10-year SCORE chart were also evaluated. We showed a pro-inflammatory/pro-oxidative milieu increasing along treatment with nilotinib compared with imatinib or dasatinib, as demonstrated by higher hs-CRP and oxLDL levels and increased IL6/IL10 and TNFα/IL10 ratios only in nilotinib cohort. After median follow-up of 23.3 months starting from TKI, 10/186 patients (5.4%) suffered an AOE. Approximately 5/10 (50%) AOEs occurred during nilotinib treatment despite a lower 10-year SCORE and a lower median age in this subgroup. A longer follow-up is needed to further confirm the active role of nilotinib in AOEs pathogenesis.
- Published
- 2022
- Full Text
- View/download PDF
29. Successful Imatinib Treatment for Systemic Mastocytosis Associated With Myelodysplastic/Myeloproliferative Neoplasm: Report of a Case and Literature Review
- Author
-
Enrico Barozzi, Cristina Bucelli, Federica Irene Grifoni, Umberto Gianelli, Alessandra Iurlo, and Daniele Cattaneo
- Subjects
imatinib ,systemic mastocytosis ,SM-AHN ,MDS/MPN ,myeloid neoplasms ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Systemic mastocytosis (SM) is a heterogeneous disease characterized by the expansion of mast cells in one or more tissues, frequently characterized by the presence of KITD816V mutation. The updated World Health Organization (WHO) classification of myeloid neoplasms recognizes SM with an associated hematological neoplasm (SM-AHN) as a new subtype among the others, which is depicted by the coexistence of SM with another hematological clonal disease. Prognosis is very different among SM patients, while its treatment, although highly personalized, is still challenging. Here we report a case of KITD816V-unmutated SM associated with MDS/MPN successfully treated with imatinib.
- Published
- 2022
- Full Text
- View/download PDF
30. Managing chronic myeloid leukemia for treatment-free remission: a proposal from the GIMEMA CML WP
- Author
-
Michele Baccarani, Elisabetta Abruzzese, Vincenzo Accurso, Francesco Albano, Mario Annunziata, Sara Barulli, Germana Beltrami, Micaela Bergamaschi, Gianni Binotto, Monica Bocchia, Giovanni Caocci, Isabella Capodanno, Francesco Cavazzini, Michele Cedrone, Marco Cerrano, Monica Crugnola, Mariella D'Adda, Chiara Elena, Carmen Fava, Paola Fazi, Claudio Fozza, Sara Galimberti, Valentina Giai, Antonella Gozzini, Gabriele Gugliotta, Alessandra Iurlo, Gaetano La Barba, Luciano Levato, Alessandro Lucchesi, Luigia Luciano, Francesca Lunghi, Monia Lunghi, Michele Malagola, Roberto Marasca, Bruno Martino, Angela Melpignano, Maria Cristina Miggiano, Enrico Montefusco, Caterina Musolino, Fausto Palmieri, Patrizia Pregno, Davide Rapezzi, Giovanna Rege-Cambrin, Serena Rupoli, Marzia Salvucci, Rosaria Sancetta, Simona Sica, Raffaele Spadano, Fabio Stagno, Mario Tiribelli, Simona Tomassetti, Elena Trabacchi, Massimiliano Bonifacio, Massimo Breccia, Fausto Castagnetti, Fabrizio Pane, Domenico Russo, Giuseppe Saglio, Simona Soverini, Paolo Vigneri, and Gianantonio Rosti
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Several papers authored by international experts have proposed recommendations on the management of BCR-ABL1+ chronic myeloid leukemia (CML). Following these recommendations, survival of CML patients has become very close to normal. The next, ambitious, step is to bring as many patients as possible into a condition of treatment-free remission (TFR). The Gruppo Italiano Malattie EMatologiche dell'Adulto (GIMEMA; Italian Group for Hematologic Diseases of the Adult) CML Working Party (WP) has developed a project aimed at selecting the treatment policies that may increase the probability of TFR, taking into account 4 variables: the need for TFR, the tyrosine kinase inhibitors (TKIs), the characteristics of leukemia, and the patient. A Delphi-like method was used to reach a consensus among the representatives of 50 centers of the CML WP. A consensus was reached on the assessment of disease risk (EUTOS Long Term Survival [ELTS] score), on the definition of the most appropriate age boundaries for the choice of first-line treatment, on the choice of the TKI for first-line treatment, and on the definition of the responses that do not require a change of the TKI (BCR-ABL1 ≤10% at 3 months, ≤1% at 6 months, ≤0.1% at 12 months, ≤0.01% at 24 months), and of the responses that require a change of the TKI, when the goal is TFR (BCR-ABL1 >10% at 3 and 6 months, >1% at 12 months, and >0.1% at 24 months). These suggestions may help optimize the treatment strategy for TFR.
- Published
- 2019
- Full Text
- View/download PDF
31. Next-generation sequencing for BCR-ABL1 kinase domain mutation testing in patients with chronic myeloid leukemia: a position paper
- Author
-
Simona Soverini, Elisabetta Abruzzese, Monica Bocchia, Massimiliano Bonifacio, Sara Galimberti, Antonella Gozzini, Alessandra Iurlo, Luigiana Luciano, Patrizia Pregno, Gianantonio Rosti, Giuseppe Saglio, Fabio Stagno, Mario Tiribelli, Paolo Vigneri, Giovanni Barosi, and Massimo Breccia
- Subjects
Next-generation sequencing ,Chronic myeloid leukemia ,Sanger sequencing ,BCR-ABL1 mutation ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract BCR-ABL1 kinase domain (KD) mutation status is considered to be an important element of clinical decision algorithms for chronic myeloid leukemia (CML) patients who do not achieve an optimal response to tyrosine kinase inhibitors (TKIs). Conventional Sanger sequencing is the method currently recommended to test BCR-ABL1 KD mutations. However, Sanger sequencing has limited sensitivity and cannot always discriminate between polyclonal and compound mutations. The use of next-generation sequencing (NGS) is increasingly widespread in diagnostic laboratories and represents an attractive alternative. Currently available data on the clinical impact of NGS-based mutational testing in CML patients do not allow recommendations with a high grade of evidence to be prepared. This article reports the results of a group discussion among an ad hoc expert panel with the objective of producing recommendations on the appropriateness of clinical decisions about the indication for NGS, the performance characteristics of NGS platforms, and the therapeutic changes that could be applied based on the use of NGS in CML. Overall, these recommendations might be employed to inform clinicians about the practical use of NGS in CML.
- Published
- 2019
- Full Text
- View/download PDF
32. Immune Dysregulation and Infectious Complications in MPN Patients Treated With JAK Inhibitors
- Author
-
Daniele Cattaneo and Alessandra Iurlo
- Subjects
myeloproliferative neoplasms ,JAK inhibitors ,ruxolitinib ,infections ,immune system ,Immunologic diseases. Allergy ,RC581-607 - Abstract
BCR-ABL1-negative myeloproliferative neoplasms are burdened by a reduced life expectancy mostly due to an increased risk of thrombo-hemorrhagic events, fibrotic progression/leukemic evolution, and infectious complications. In these clonal myeloid malignancies, JAK2V617F is the main driver mutation, leading to an aberrant activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway. Therefore, its inhibition represents an attractive therapeutic strategy for these disorders. Several JAK inhibitors have entered clinical trials, including ruxolitinib, the first JAK1/2 inhibitor to become commercially available for the treatment of myelofibrosis and polycythemia vera. Due to interference with the JAK-STAT pathway, JAK inhibitors affect several components of the innate and adaptive immune systems such as dendritic cells, natural killer cells, T helper cells, and regulatory T cells. Therefore, even though the clinical use of these drugs in MPN patients has led to a dramatic improvement of symptoms control, organ involvement, and quality of life, JAK inhibitors–related loss of function in JAK-STAT signaling pathway can be a cause of different adverse events, including those related to a condition of immune suppression or deficiency. This review article will provide a comprehensive overview of the current knowledge on JAK inhibitors’ effects on immune cells as well as their clinical consequences, particularly with regards to infectious complications.
- Published
- 2021
- Full Text
- View/download PDF
33. Co-Occurrence of Myeloid and Lymphoid Neoplasms: Clinical Characterization and Impact on Outcome. A Single-Center Cohort Study
- Author
-
Cristina Bucelli, Bruno Fattizzo, Daniele Cattaneo, Juri Alessandro Giannotta, Kordelia Barbullushi, Raffaella Pasquale, Enrico Barozzi, Maria Chiara Barbanti, Loredana Pettine, Francesca Gaia Rossi, Gianluigi Reda, Ramona Cassin, Wilma Barcellini, Luca Baldini, and Alessandra Iurlo
- Subjects
myeloproliferative neoplasms ,lymphoproliferative syndromes ,myelodysplastic syndromes ,infections ,secondary malignancies ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The co-occurrence of myeloid neoplasms and lymphoproliferative diseases (LPDs) has been epidemiologically described, particularly in myeloproliferative neoplasms (MPNs). However, the clinical features of these patients are poorly known. In this study, we evaluated a single-center cohort of 44 patients with a diagnosis of myeloid and LPD focusing on clinical features, therapy requirement, and outcome. The two diagnoses were concomitant in 32% of patients, while myeloid disease preceded LPD in 52% of cases (after a median of 37 months, 6–318), and LPD preceded myeloid neoplasm in 16% (after a median of 41 months, 5–242). The most prevalent LPD was non-Hodgkin lymphoma (50%), particularly lymphoplasmacytic lymphoma (54.5%), followed by chronic lymphocytic leukemia (27%), plasma cell dyscrasias (18.2%), and rarer associations such as Hodgkin lymphoma and Erdheim–Chester disease. Overall, 80% of BCR-ABL1-negative MPN patients required a myeloid-specific treatment and LPD received therapy in 45.5% of cases. Seven subjects experienced vascular events, 13 a grade >/= 3 infectious episode (9 pneumonias, 3 urinary tract infection, and 1 sepsis), and 9 developed a solid tumor. Finally, nine patients died due to solid tumor (four), leukemic progression (two), infectious complications (two), and brain bleeding (one). Longer survival was observed in younger patients (p = 0.001), with better performance status (p = 0.02) and in the presence of driver mutations (p = 0.003). Contrarily, a worse survival was significantly associated with the occurrence of infections (p < 0.0001). These data suggest that in subjects with co-occurrence of myeloid and lymphoid neoplasms, high medical surveillance for infectious complications is needed, along with patient education, since they may negatively impact outcome.
- Published
- 2021
- Full Text
- View/download PDF
34. Second primary malignancies in ruxolitinib-treated myelofibrosis: real-world evidence from 219 consecutive patients
- Author
-
Margherita Maffioli, Toni Giorgino, Barbara Mora, Alessandra Iurlo, Elena Elli, Maria Chiara Finazzi, Marianna Caramella, Elisa Rumi, Maria Cristina Carraro, Nicola Polverelli, Mariella D'Adda, Simona Malato, Marianna Rossi, Alfredo Molteni, Alessandro Vismara, Cinzia Sissa, Francesco Spina, Michela Anghilieri, Daniele Cattaneo, Rossella Renso, Marta Bellini, Maria Luisa Pioltelli, Chiara Cavalloni, Daniela Barraco, Raffaella Accetta, Lorenza Bertù, Matteo Giovanni Della Porta, and Francesco Passamonti
- Subjects
Specialties of internal medicine ,RC581-951 - Published
- 2019
- Full Text
- View/download PDF
35. Digital PCR improves the quantitation of DMR and the selection of CML candidates to TKIs discontinuation
- Author
-
Simona Bernardi, Michele Malagola, Camilla Zanaglio, Nicola Polverelli, Elif Dereli Eke, Mariella D’Adda, Mirko Farina, Cristina Bucelli, Luigi Scaffidi, Eleonora Toffoletti, Clara Deambrogi, Fabio Stagno, Micaela Bergamaschi, Luca Franceschini, Elisabetta Abruzzese, Maria Domenica Divona, Marco Gobbi, Francesco Di Raimondo, Gianluca Gaidano, Mario Tiribelli, Massimiliano Bonifacio, Chiara Cattaneo, Alessandra Iurlo, and Domenico Russo
- Subjects
chronic myeloid leukemia ,digital PCR (dPCR) ,minimal residual disease (MRD) monitoring ,treatment‐free remission (TFR) ,tyrosine kinase inhibitors (TKI) discontinuation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Treatment‐free remission (TFR) by tyrosine kinase inhibitors (TKI) discontinuation in patients with deep molecular response (DMR) is a paramount goal in the current chronic myeloid leukemia (CML) therapeutic strategy. The best DMR level by real‐time quantitative PCR (RT‐qPCR) for TKI discontinuation is still a matter of debate. To compare the accuracy of digital PCR (dPCR) and RT‐qPCR for BCR‐ABL1 transcript levels detection, 142 CML patients were monitored for a median time of 24 months. Digital PCR detected BCR‐ABL1 transcripts in the RT‐qPCR undetectable cases. The dPCR analysis of the samples, grouped by the MR classes, revealed a significant difference between MR4.0 and MR4.5 (P = 0.0104) or MR5.0 (P = 0.0032). The clinical and hematological characteristics of the patients grouped according to DMR classes (MR4.0 vs MR4.5‐5.0) were superimposable. Conversely, patients with dPCR values
- Published
- 2019
- Full Text
- View/download PDF
36. Perspectives and Emotional Experiences of Patients With Chronic Myeloid Leukemia During ENESTPath Clinical Trial and Treatment-Free Remission: Rationale and Protocol of the Italian Substudy
- Author
-
Lidia Borghi, Gianantonio Rosti, Alessandro Maggi, Massimo Breccia, Eros Di Bona, Alessandra Iurlo, Gaetano La Barba, Paolo Sportoletti, Francesco Albano, Sara Galimberti, Flavia Rivellini, Giovanna Rege Cambrin, Isabella Capodanno, Antonio Cuneo, Massimiliano Bonifacio, Simona Sica, Luca Arcaini, Enrico Capochiani, Claudia Minotto, Fabio Ciceri, Monica Crugnola, Luigi Di Caprio, Sharon Supekar, Chiara Elena, Michele Baccarani, and Elena Vegni
- Subjects
Chronic myeloid leukemia ,ENESTPath ,emotional experience ,nilotinib ,psychological distress ,quality of life ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Achievement of deep molecular response following treatment with a tyrosine kinase inhibitor (TKI) allows for treatment-free remission (TFR) in many patients with chronic myeloid leukemia (CML). Successful TFR is defined as the achievement of a sustained molecular response after cessation of ongoing TKI therapy. The phase 3 ENESTPath study was designed to determine the required optimal duration of consolidation treatment with the second-generation TKI, nilotinib 300 mg twice-daily, to remain in successful TFR without relapse after entering TFR for 12 months. The purpose of this Italian ‘patient’s voice CML’ substudy was to evaluate patients’ psycho-emotional characteristics and quality of life through their experiences of stopping treatment with nilotinib and entering TFR. The purpose of the present contribution is to early present the study protocol of an ongoing study to the scientific community, in order to describe the study rationale and to extensively present the study methodology. Patients aged ≥18 years with a confirmed diagnosis of Philadelphia chromosome positive BCR-ABL1+ CML in chronic phase and treated with front-line imatinib for a minimum of 24 months from the enrollment were eligible. Patients consenting to participate the substudy will have quality of life questionnaires and in-depth qualitative interviews conducted. The substudy will include both qualitative and quantitative design aspects to evaluate the psychological outcomes as assessed via patients’ emotional experience during and after stopping nilotinib therapy. Randomization is hypothesized to be a timepoint of higher psychological alert or distress when compared to consolidation and additionally any improvement in health-related quality of life (HRQoL) due to nilotinib treatment is expected across the timepoints (from consolidation, to randomization, and TFR). An association is also expected between dysfunctional coping strategies, such as detachments and certain personality traits, and psychological distress and HRQoL impairments. Better HRQoL outcomes are expected in TFR compared to the end of consolidation. This substudy is designed for in-depth assessment of all potential psycho-emotional variables and aims to determine the need for personalized patient care and counselling, and also guide clinicians to consider the psychological well-being of patients who are considering treatment termination.NCT number: NCT01743989, EudraCT number: 2012-005124-15
- Published
- 2021
- Full Text
- View/download PDF
37. Triple-Negative Essential Thrombocythemia: Clinical-Pathological and Molecular Features. A Single-Center Cohort Study
- Author
-
Daniele Cattaneo, Giorgio Alberto Croci, Cristina Bucelli, Silvia Tabano, Marta Giulia Cannone, Gabriella Gaudioso, Maria Chiara Barbanti, Kordelia Barbullushi, Paola Bianchi, Elisa Fermo, Sonia Fabris, Luca Baldini, Umberto Gianelli, and Alessandra Iurlo
- Subjects
essential thrombocytemia ,triple-negative ,bone marrow morphology ,next-generation sequencing ,prognosis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Lack of demonstrable mutations affecting JAK2, CALR, or MPL driver genes within the spectrum of BCR-ABL1-negative myeloproliferative neoplasms (MPNs) is currently referred to as a triple-negative genotype, which is found in about 10% of patients with essential thrombocythemia (ET) and 5–10% of those with primary myelofibrosis (PMF). Very few papers are presently available on triple-negative ET, which is basically described as an indolent disease, differently from triple-negative PMF, which is an aggressive myeloid neoplasm, with a significantly higher risk of leukemic evolution. The aim of the present study was to evaluate the bone marrow morphology and the clinical-laboratory parameters of triple-negative ET patients, as well as to determine their molecular profile using next-generation sequencing (NGS) to identify any potential clonal biomarkers. We evaluated a single-center series of 40 triple-negative ET patients, diagnosed according to the 2017 WHO classification criteria and regularly followed up at the Hematology Unit of our Institution, between January 1983 and January 2019. In all patients, NGS was performed using the Illumina Ampliseq Myeloid Panel; morphological and immunohistochemical features of the bone marrow trephine biopsies were also thoroughly reviewed. Nucleotide variants were detected in 35 out of 40 patients. In detail, 29 subjects harbored one or two variants and six cases showed three or more concomitant nucleotide changes. The most frequent sequence variants involved the TET2 gene (55.0%), followed by KIT (27.5%). Histologically, most of the cases displayed a classical ET morphology. Interestingly, prevalent megakaryocytes morphology was more frequently polymorphic with a mixture of giant megakaryocytes with hyperlobulated nuclei, normal and small sized maturing elements, and naked nuclei. Finally, in five cases a mild degree of reticulin fibrosis (MF-1) was evident together with an increase in the micro-vessel density. By means of NGS we were able to identify nucleotide variants in most cases, thus we suggest that a sizeable proportion of triple-negative ET patients do have a clonal disease. In analogy with driver genes-mutated MPNs, these observations may prevent issues arising concerning triple-negative ET treatment, especially when a cytoreductive therapy may be warranted.
- Published
- 2021
- Full Text
- View/download PDF
38. Current Strategies and Future Directions to Achieve Deep Molecular Response and Treatment-Free Remission in Chronic Myeloid Leukemia
- Author
-
Mario Annunziata, Massimiliano Bonifacio, Massimo Breccia, Fausto Castagnetti, Antonella Gozzini, Alessandra Iurlo, Patrizia Pregno, Fabio Stagno, and Giorgina Specchia
- Subjects
chronic myeloid leukemia ,deep molecular response ,optimal strategies ,treatment-free remission ,tyrosine kinase inhibitors ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The treatment of chronic myeloid leukemia (CML) has been radically changed by the approval of tyrosine kinase inhibitors (TKIs), which target BCR-ABL1 kinase activity. CML is now managed as a chronic disease requiring long-term treatment and close molecular monitoring. It has been shown that in a substantial number of patients who have achieved a stable deep molecular response (DMR), TKI treatment can be safely discontinued without loss of response. Therefore, treatment-free remission (TFR), through the achievement of a DMR, is increasingly regarded as a feasible treatment goal in many CML patients. However, only nilotinib has approval in this setting and a number of controversial aspects remain regarding treatment choices and timings, predictive factors, patient communication, and optimal strategies to achieve successful TFR. This narrative review aims to provide a comprehensive overview on how to optimize the path to DMR and TFR in patients with CML, and discusses recent data and future directions.
- Published
- 2020
- Full Text
- View/download PDF
39. Disease progression in myeloproliferative neoplasms: comparing patients in accelerated phase with those in chronic phase with increased blasts
- Author
-
Julia T. Geyer, Elizabeth Margolskee, Spencer A. Krichevsky, Daniele Cattaneo, Leonardo Boiocchi, Paola Ronchi, Francesca Lunghi, Joseph M. Scandura, Maurilio Ponzoni, Robert P. Hasserjian, Umberto Gianelli, Alessandra Iurlo, and Attilio Orazi
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
40. An unusual type of myeloid sarcoma localization following myelofibrosis: A case report and literature review
- Author
-
Nicola Orofino, Daniele Cattaneo, Cristina Bucelli, Loredana Pettine, Sonia Fabris, Umberto Gianelli, Nicola Stefano Fracchiolla, Agostino Cortelezzi, and Alessandra Iurlo
- Subjects
Myeloid sarcoma ,Primary myelofibrosis ,Calreticulin ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Myeloid Sarcoma (MS) is a rare malignancy that can present as an isolated disease or more frequently in association with or following acute myeloid leukemia or other myeloid neoplasms and rarely following myelofibrosis. Since molecular pathogenesis and prognostic factors of MS are not well understood, its prognosis remains poor even in the era of novel agents and target therapies. We report the case of a patient with MS following myelofibrosis with multiple subcutaneous, cutaneous and muscle localizations; the latter has been reported in the literature as anecdotal. In this way we aimed to enhance the understanding of this disease.
- Published
- 2017
- Full Text
- View/download PDF
41. Observational study of chronic myeloid leukemia Italian patients who discontinued tyrosine kinase inhibitors in clinical practice
- Author
-
Carmen Fava, Giovanna Rege-Cambrin, Irene Dogliotti, Marco Cerrano, Paola Berchialla, Matteo Dragani, Gianantonio Rosti, Fausto Castagnetti, Gabriele Gugliotta, Bruno Martino, Carlo Gambacorti-Passerini, Elisabetta Abruzzese, Chiara Elena, Patrizia Pregno, Antonella Gozzini, Isabella Capodanno, Micaela Bergamaschi, Monica Crugnola, Monica Bocchia, Sara Galimberti, Davide Rapezzi, Alessandra Iurlo, Daniele Cattaneo, Roberto Latagliata, Massimo Breccia, Michele Cedrone, Marco Santoro, Mario Annunziata, Luciano Levato, Fabio Stagno, Francesco Cavazzini, Nicola Sgherza, Valentina Giai, Luigia Luciano, Sabina Russo, Pellegrino Musto, Giovanni Caocci, Federica Sorà, Francesco Iuliano, Francesca Lunghi, Giorgina Specchia, Fabrizio Pane, Dario Ferrero, Michele Baccarani, and Giuseppe Saglio
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
It is judged safe to discontinue treatment with tyrosine kinase inhibitors (TKI) for chronic myeloid leukemia (CML) in experimental trials on treatment-free remission (TFR). We collected a total of 293 Italian patients with chronic phase CML who discontinued TKI in deep molecular response. Seventy-two percent of patients were on treatment with imatinib, and 28% with second generation TKI at the time of discontinuation. Median duration of treatment with the last TKI was 77 months [Interquartile Range (IQR) 54;111], median duration of deep molecular response was 46 months (IQR 31;74). Duration of treatment with TKI and duration of deep molecular response were shorter with second generation TKI than with imatinib (P
- Published
- 2019
- Full Text
- View/download PDF
42. The Role of New Technologies in Myeloproliferative Neoplasms
- Author
-
Giuseppe A. Palumbo, Stefania Stella, Maria Stella Pennisi, Cristina Pirosa, Elisa Fermo, Sonia Fabris, Daniele Cattaneo, and Alessandra Iurlo
- Subjects
BCR-ABL1-negative myeloproliferative neoplasms (MPNs) ,myelofibrosis (MF) ,JAK2 mutations ,calreticulin (CALR) ,MPL (W515K/L) ,ASXL1 mutation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The hallmark of BCR-ABL1-negative myeloproliferative neoplasms (MPNs) is the presence of a driver mutation in JAK2, CALR, or MPL gene. These genetic alterations represent a key feature, useful for diagnostic, prognostic and therapeutical approaches. Molecular biology tests are now widely available with different specificity and sensitivity. Recently, the allele burden quantification of driver mutations has become a useful tool, both for prognostication and efficacy evaluation of therapies. Moreover, other sub-clonal mutations have been reported in MPN patients, which are associated with poorer prognosis. ASXL1 mutation appears to be the worst amongst them. Both driver and sub-clonal mutations are now taken into consideration in new prognostic scoring systems and may be better investigated using next generation sequence (NGS) technology. In this review we summarize the value of NGS and its contribution in providing a comprehensive picture of mutational landscape to guide treatment decisions. Finally, discussing the role that NGS has in defining the potential risk of disease development, we forecast NGS as the standard molecular biology technique for evaluating these patients.
- Published
- 2019
- Full Text
- View/download PDF
43. Chronic Myeloid Leukemia Patient’s Voice About the Experience of Treatment-Free Remission Failure: Results From the Italian Sub-Study of ENESTPath Exploring the Emotional Experience of Patients During Different Phases of a Clinical Trial
- Author
-
Lidia Borghi, Sara Galimberti, Claudia Baratè, Massimiliano Bonifacio, Enrico Capochiani, Antonio Cuneo, Franca Falzetti, Alessandra Iurlo, Francesca Lunghi, Claudia Minotto, Ester Maria Orlandi, Giovanna Rege-Cambrin, Simona Sica, Sharon Supekar, Jens Haenig, and Elena Vegni
- Subjects
chronic myeloid leukemia ,nilotinib ,ENESTPath ,TFR ,inner emotional experience ,psycho-emotional outcomes ,Psychology ,BF1-990 - Abstract
Background: The main objective of this study is to gain further insights on how chronic myeloid leukemia (CML) patients involved in an interventional clinical trial with the purpose of reaching treatment free remission (TFR) phase, perceived and experienced TFR failure. TFR failure was defined for the individual patient as either not being eligible for drug discontinuation or as having relapse in the TFR phase with reintroduction of nilotinib treatment.Methods: Using a qualitative approach, out of 25 patients with CML who experienced TFR failure 14 were interviewed. Patients’ views and experiences were explored using in-depth interviews, analyzed using the Interpretative Phenomenological Analysis (IPA).Results: The analysis of the interviews revealed that the experience of the diagnosis seems to have been lived as a traumatic break that has created a dichotomy, like an ambivalence in the ways in which CML patients perceived and experienced the whole disease journey, with contradictory feelings of both positive and negative emotions (e.g., a diagnosis of cancer, that is something distressing and of being afraid of, but also with a treatment and a life expectancies of which being grateful). This ambivalence of feelings was found to give meaning to the way in which patients cognitively and emotionally experienced the different steps of their disease history. Thus, four main issues, corresponding to different steps of the patients’ journey, were identified: (1) the moment of the diagnosis, (2) the experience of the illness journey: disease and treatment, (3) the moment of “TFR failure,” and (4) the impact of disease, treatment and relapse on the patient’s life.Conclusion: This qualitative analysis helps in understanding patients’ perspective, both in terms of getting access to the inner subjective experience of having CML and its strict relationship with the involvement in a trial or its cessation. Clinicians should consider that the way in which CML patients feel engaged in a clinical trial, create expectancies about TFR or experience the TFR failure is linked to the process of coping with the diagnosis, which is characterized by ambivalence.
- Published
- 2019
- Full Text
- View/download PDF
44. Nilotinib interferes with cell cycle, ABC transporters and JAK-STAT signaling pathway in CD34+/lin- cells of patients with chronic phase chronic myeloid leukemia after 12 months of treatment.
- Author
-
Alessandra Trojani, Ester Pungolino, Alessandra Dal Molin, Milena Lodola, Giuseppe Rossi, Mariella D'Adda, Alessandra Perego, Chiara Elena, Mauro Turrini, Lorenza Borin, Cristina Bucelli, Simona Malato, Maria Cristina Carraro, Francesco Spina, Maria Luisa Latargia, Salvatore Artale, Pierangelo Spedini, Michela Anghilieri, Barbara Di Camillo, Giacomo Baruzzo, Gabriella De Canal, Alessandra Iurlo, Enrica Morra, and Roberto Cairoli
- Subjects
Medicine ,Science - Abstract
Chronic myeloid leukemia (CML) is characterized by the constitutive tyrosine kinase activity of the oncoprotein BCR-ABL1 in myeloid progenitor cells that activates multiple signal transduction pathways leading to the leukemic phenotype. The tyrosine-kinase inhibitor (TKI) nilotinib inhibits the tyrosine kinase activity of BCR-ABL1 in CML patients. Despite the success of nilotinib treatment in patients with chronic-phase (CP) CML, a population of Philadelphia-positive (Ph+) quiescent stem cells escapes the drug activity and can lead to drug resistance. The molecular mechanism by which these quiescent cells remain insensitive is poorly understood. The aim of this study was to compare the gene expression profiling (GEP) of bone marrow (BM) CD34+/lin- cells from CP-CML patients at diagnosis and after 12 months of nilotinib treatment by microarray, in order to identify gene expression changes and the dysregulation of pathways due to nilotinib action. We selected BM CD34+/lin- cells from 78 CP-CML patients at diagnosis and after 12 months of first-line nilotinib therapy and microarray analysis was performed. GEP bioinformatic analyses identified 2,959 differently expressed probes and functional clustering determined some significantly enriched pathways between diagnosis and 12 months of nilotinib treatment. Among these pathways, we observed the under expression of 26 genes encoding proteins belonging to the cell cycle after 12 months of nilotinib treatment which led to the up-regulation of chromosome replication, cell proliferation, DNA replication, and DNA damage checkpoint at diagnosis. We demonstrated the under expression of the ATP-binding cassette (ABC) transporters ABCC4, ABCC5, and ABCD3 encoding proteins which pumped drugs out of the cells after 12 months of nilotinib. Moreover, GEP data demonstrated the deregulation of genes involved in the JAK-STAT signaling pathway. The down-regulation of JAK2, IL7, STAM, PIK3CA, PTPN11, RAF1, and SOS1 key genes after 12 months of nilotinib could demonstrate the up-regulation of cell cycle, proliferation and differentiation via MAPK and PI3K-AKT signaling pathways at diagnosis.
- Published
- 2019
- Full Text
- View/download PDF
45. The Polycomb BMI1 Protein Is Co-expressed With CD26+ in Leukemic Stem Cells of Chronic Myeloid Leukemia
- Author
-
Sara Galimberti, Susanna Grassi, Claudia Baratè, Francesca Guerrini, Elena Ciabatti, Francesca Perutelli, Federica Ricci, Giada Del Genio, Marina Montali, Serena Barachini, Cecilia Giuliani, Maria Immacolata Ferreri, Angelo Valetto, Elisabetta Abruzzese, Chiara Ippolito, Alessandra Iurlo, Monica Bocchia, Anna Sicuranza, Bruno Martino, Lorenzo Iovino, Gabriele Buda, Serena Salehzadeh, Mario Petrini, Antonello Di Paolo, and Letizia Mattii
- Subjects
BMI1 ,polycomb ,BCR-ABL1 ,CML ,CD26 ,leukemic stem cell ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The Polycomb gene BMI1 expression exerts a negative predictive impact on several hematological malignancies, such as acute and chronic myeloid leukemia (CML), myelofibrosis, and follicular lymphoma. As already demonstrated in CML, BMI1 is responsible for the resistance to the tyrosine kinase inhibitors (TKIs) in a BCR-ABL1-independent way. Even if, it is unknown where BMI1 in CML is expressed (in progenitors or more mature cells). We decided, therefore, to evaluate if and where the BMI1 protein is located, focusing mainly on the CD34+/CD38-/CD26+ CML progenitors. To begin we measured, by flow cytometry, the proportion of CD34+/CD26+ cells in 31 bone marrow samples from 20 CML patients, at diagnosis and during treatment with imatinib. After that the bone marrow blood smears were stained with antibodies anti-CD26, BCR-ABL1, and BMI1. These smears were observed by a confocal laser microscope and a 3D reconstruction was then performed. At diagnosis, CD34+/CD26+ cells median value/μL was 0.48; this number increased from diagnosis to the third month of therapy and then reduced during treatment with imatinib. The number and behavior of the CD26+ progenitors were independent from the BCR-ABL1 expression, but they summed up what previously observed about the BMI1 expression modulation. In this work we demonstrate for the first time that in CML the BMI1 protein is co-expressed with BCR-ABL1 only in the cytoplasm of the CD26+ precursors; on the contrary, in other hematological malignancies where BMI1 is commonly expressed (follicular lymphoma, essential thrombocytemia, acute myeloid leukemia), it was not co-localized with CD26 or, obviously, with BCR-ABL1. Once translated into the clinical context, if BMI1 is a marker of stemness, our results would suggest the combination of the BMI1 inhibitors with TKIs as an interesting object of research, and, probably, as a promising way to overcome resistance in CML patients.
- Published
- 2018
- Full Text
- View/download PDF
46. HHV8-Negative Effusion-Based Large B Cell Lymphoma Arising in Chronic Myeloid Leukemia Patients under Dasatinib Treatment: A Report of Two Cases
- Author
-
Stefano Fiori, Elisabetta Todisco, Safaa Ramadan, Federica Gigli, Patrizia Falco, Alessandra Iurlo, Cristiano Rampinelli, Giorgio Croci, Stefano A. Pileri, and Corrado Tarella
- Subjects
CML ,TKIs ,large B cell lymphoma ,EBL ,pleural effusion ,dasatinib ,Biology (General) ,QH301-705.5 - Abstract
Tyrosine kinase inhibitors (TKIs) are the treatment of choice for BCR-ABL1-positive chronic myeloid leukemia (CML). Although TKIs have substantially improved prognosis of CML patients, their use is not free of adverse effects. Dasatinib is a second generation TKI frequently associated with pleural effusion in up to 33% of patients. This results in symptoms as dyspnea, cough and chest pain that may require therapy discontinuation. In the present report, we describe two exceptional cases of HHV8-negative large B-cell effusion-based lymphoma (EBL) confined to the pleura, incidentally, diagnosed in patients presenting with dasatinib-related pleural effusion. One patient (case 1) is alive and is in remission at 17 months from large B-cell EBL diagnosis while unfortunately the other patient (case 2) died of progressive disease and COVID-19 pneumonia 16 months from large B-cell EBL diagnosis. These cases raise concern about a possible association between large B-cell EBL and dasatinib, and the different clinical outcome of the two cases poses a challenge in treatment decision. For this reason, we strongly recommend cytological investigation in patients with persistent/relapsing pleural effusion under dasatinib, primarily to validate its possible association with lymphoma development and to improve the knowledge about this entity.
- Published
- 2021
- Full Text
- View/download PDF
47. Frontline Dasatinib Treatment in a 'Real-Life' Cohort of Patients Older than 65 Years with Chronic Myeloid Leukemia
- Author
-
Roberto Latagliata, Fabio Stagno, Mario Annunziata, Elisabetta Abruzzese, Alessandra Iurlo, Attilio Guarini, Carmen Fava, Antonella Gozzini, Massimiliano Bonifacio, Federica Sorà, Sabrina Leonetti Crescenzi, Monica Bocchia, Monica Crugnola, Fausto Castagnetti, Isabella Capodanno, Sara Galimberti, Costanzo Feo, Raffaele Porrini, Patrizia Pregno, Manuela Rizzo, Agostino Antolino, Endri Mauro, Nicola Sgherza, Luigiana Luciano, Mario Tiribelli, Antonella Russo Rossi, Malgorzata Trawinska, Paolo Vigneri, Massimo Breccia, Gianantonio Rosti, and Giuliana Alimena
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Dasatinib (DAS) has been licensed for the frontline treatment in chronic myeloid leukemia (CML). However, very few data are available regarding its efficacy and toxicity in elderly patients with CML outside clinical trials. To address this issue, we set out a “real-life” cohort of 65 chronic phase CML patients older than 65 years (median age 75.1 years) treated frontline with DAS in 26 Italian centers from June 2012 to June 2015, focusing our attention on toxicity and efficacy data. One third of patients (20/65: 30.7%) had 3 or more comorbidities and required concomitant therapies; according to Sokal classification, 3 patients (4.6%) were low risk, 39 (60.0%) intermediate risk, and 20 (30.8%) high risk, whereas 3 (4.6%) were not classifiable. DAS starting dose was 100 mg once a day in 54 patients (83.0%), whereas 11 patients (17.0%) received less than 100 mg/day. Grade 3/4 hematologic and extrahematologic toxicities were reported in 8 (12.3%) and 12 (18.5%) patients, respectively. Overall, 10 patients (15.4%) permanently discontinued DAS because of toxicities. Pleural effusions (all WHO grades) occurred in 12 patients (18.5%) and in 5 of them occurred during the first 3 months. DAS treatment induced in 60/65 patients (92.3%) a complete cytogenetic response and in 50/65 (76.9%) also a major molecular response. These findings show that DAS might play an important role in the frontline treatment of CML patients >65 years old, proving efficacy and having a favorable safety profile also in elderly subjects with comorbidities.
- Published
- 2016
- Full Text
- View/download PDF
48. Residual Peripheral Blood CD26+ Leukemic Stem Cells in Chronic Myeloid Leukemia Patients During TKI Therapy and During Treatment-Free Remission
- Author
-
Monica Bocchia, Anna Sicuranza, Elisabetta Abruzzese, Alessandra Iurlo, Santina Sirianni, Antonella Gozzini, Sara Galimberti, Lara Aprile, Bruno Martino, Patrizia Pregno, Federica Sorà, Giulia Alunni, Carmen Fava, Fausto Castagnetti, Luca Puccetti, Massimo Breccia, Daniele Cattaneo, Marzia Defina, Olga Mulas, Claudia Baratè, Giovanni Caocci, Simona Sica, Alessandro Gozzetti, Luigiana Luciano, Monica Crugnola, Mario Annunziata, Mario Tiribelli, Paola Pacelli, Ilaria Ferrigno, Emilio Usala, Nicola Sgherza, Gianantonio Rosti, Alberto Bosi, and Donatella Raspadori
- Subjects
chronic myeloid leukemia ,CD26 ,leukemic stem cells ,TKI ,minimal residual disease ,flow cytometry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Chronic myeloid leukemia (CML) patients in sustained “deep molecular response” may stop TKI treatment without disease recurrence; however, half of them lose molecular response shortly after TKI withdrawing. Well-defined eligibility criteria to predict a safe discontinuation up-front are still missing. Relapse is probably due to residual quiescent TKI-resistant leukemic stem cells (LSCs) supposedly transcriptionally low/silent and not easily detectable by BCR-ABL1 qRT-PCR. Bone marrow Ph+ CML CD34+/CD38− LSCs were found to specifically co-express CD26 (dipeptidylpeptidase-IV). We explored feasibility of detecting and quantifying CD26+ LSCs by flow cytometry in peripheral blood (PB). Over 400 CML patients (at diagnosis and during/after therapy) entered this cross-sectional study in which CD26 expression was evaluated by a standardized multiparametric flow cytometry analysis on PB CD45+/CD34+/CD38− stem cell population. All 120 CP-CML patients at diagnosis showed measurable PB CD26+ LSCs (median 19.20/μL, range 0.27–698.6). PB CD26+ LSCs were also detectable in 169/236 (71.6%) CP-CML patients in first-line TKI treatment (median 0.014 cells/μL; range 0.0012–0.66) and in 74/112 (66%), additional patients studied on treatment-free remission (TFR) (median 0.015/μL; range 0.006–0.76). Notably, no correlation between BCR-ABL/ABLIS ratio and number of residual LSCs was found both in patients on or off TKIs. This is the first evidence that “circulating” CML LSCs persist in the majority of CML patients in molecular response while on TKI treatment and even after TKI discontinuation. Prospective studies evaluating the dynamics of PB CD26+ LSCs during TKI treatment and the role of a “stem cell response” threshold to achieve and maintain TFR are ongoing.
- Published
- 2018
- Full Text
- View/download PDF
49. Early switch to nilotinib in a case of non-optimal response to imatinib
- Author
-
Alessandra Iurlo, Tommaso Radice, Chiara De Philippis, Manuela Zappa, Mauro Pomati, and Agostino Cortelezzi
- Subjects
Chronic Myeloid Leukemia ,Early switch ,Nilotinib ,Medicine (General) ,R5-920 - Abstract
We report a case of excellent response to nilotinib in a 22 years old man with chronic myeloid leukemia in suboptimal response to imatinib. After diagnosis he started cytoreductive therapy with cytarabine and hydroxyurea, then he begun therapy with imatinib 400 mg/day. After 3 months of treatment, he obtained a complete hematologic response (CHR) and a minor cytogenetic response (minor CyR). At 6 months CHR was confirmed, but bone marrow analysis showed increasing number of Ph+ cells (minimal CyR) and non significant reduction of BCR-ABL levels. According to ELN (European LeukemiaNet) guidelines, this is considered a suboptimal response. Clonal evolution, kinase domain mutations and reduced drug intake were excluded, thus we decided to early switch to nilotinib at 400 mg/BID. After 3 months of treatment we obtained a complete cytogenetic response (CCyR) and a strong reduction of BCR-ABL transcript, almost reaching a major molecular response (MMR).
- Published
- 2015
- Full Text
- View/download PDF
50. Incidence of second primary malignancies and related mortality in patients with imatinib-treated chronic myeloid leukemia
- Author
-
Gabriele Gugliotta, Fausto Castagnetti, Massimo Breccia, Francesco Albano, Alessandra Iurlo, Tamara Intermesoli, Elisabetta Abruzzese, Luciano Levato, Mariella D’Adda, Patrizia Pregno, Francesco Cavazzini, Fabio Stagno, Bruno Martino, Gaetano La Barba, Federica Sorà, Mario Tiribelli, Catia Bigazzi, Gianni Binotto, Massimiliano Bonifacio, Clementina Caracciolo, Simona Soverini, Robin Foà, Michele Cavo, Giovanni Martinelli, Fabrizio Pane, Giuseppe Saglio, Michele Baccarani, and Gianantonio Rosti
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
The majority of patients with chronic myeloid leukemia are successfully managed with life-long treatment with tyrosine kinase inhibitors. In patients in chronic phase, other malignancies are among the most common causes of death, raising concerns on the relationship between these deaths and the off-target effects of tyrosine kinase inhibitors. We analyzed the incidence of second primary malignancies, and related mortality, in 514 chronic myeloid leukemia patients enrolled in clinical trials in which imatinib was given as first-line treatment. We then compared the observed incidence and mortality with those expected in the age- and sex-matched Italian general population, calculating standardized incidence and standardized mortality ratios. After a median follow-up of 74 months, 5.8% patients developed second primary malignancies. The median time from chronic myeloid leukemia to diagnosis of the second primary malignancies was 34 months. We did not find a higher incidence of second primary malignancies compared to that in the age- and sex-matched Italian general population, with standardized incidence ratios of 1.06 (95% CI: 0.57–1.54) and 1.61 (95% CI: 0.92–2.31) in males and females, respectively. Overall, 3.1% patients died of second primary malignancies. The death rate in patients with second primary malignancies was 53% (median overall survival: 18 months). Among females, the observed cancer-related mortality was superior to that expected in the age- and sex-matched Italian population, with a standardized mortality ratio of 2.41 (95% CI: 1.26 – 3.56). In conclusion, our analysis of patients with imatinib-treated chronic myeloid leukemia did not reveal a higher incidence of second primary malignancies; however, the outcome of second primary malignancies in such patients was worse than expected. Clinicaltrials.gov: NCT00514488, NCT00510926.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.