Ciavarella, S., Vegliante, M.C., Fabbri, M., De Summa, S., Melle, F., Motta, G., De Iuliis, V., Opinto, G., Enjuanes, A., Rega, S., Gulino, A., Agostinelli, C., Scattone, A., Tommasi, S., Mangia, A., Mele, F., Simone, G., Zito, A.F., Ingravallo, G., Vitolo, U., Chiappella, A., Tarella, C., Gianni, A.M., Rambaldi, A., Zinzani, P.L., Casadei, B., Derenzini, E., Loseto, G., Pileri, A., Tabanelli, V., Fiori, S., Rivas-Delgado, A., López-Guillermo, A., Venesio, T., Sapino, A., Campo, E., Tripodo, C., Guarini, A., and Pileri, S.A.
Maraglino, A. M. E., Amato, V., Sammassimo, S., Gigli, F., Tabanelli, V., Pastano, R., Tarella, C., Giglio, F., and Derenzini, E.
Subjects
CHRONIC leukemia, GRAFT versus host disease
Abstract
HT
Patients affected by CMML with NEMMs
Patients with AML transformation
Patients, total number
7 All studies included in this analysis describe patients affected by CMML with histologically confirmed malignant NEMMs (defined by the presence of malignant CMML cells) occurring at any time during the disease course. NEMMs were documented in 47 patients (75%) within 12 months from initial CMML diagnosis and in 16 patients (25%) later in the disease course. The patient's cohort analyzed in this study includes 77 patients with malignant NEMMs from the published series and our case-report (78 patients in total). [Extracted from the article]
Derenzini E, Mazzara S, Melle F, Motta G, Fabbri M, Bruna R, Agostinelli C, Cesano A, Corsini CA, Chen N, Righi S, Sabattini E, Chiappella A, Calleri A, Fiori S, Tabanelli V, Cabras A, Pruneri G, Vitolo U, Gianni AM, Rambaldi A, Corradini P, Zinzani PL, Tarella C, Pileri S., and Derenzini E, Mazzara S, Melle F, Motta G, Fabbri M, Bruna R, Agostinelli C, Cesano A, Corsini CA, Chen N, Righi S, Sabattini E, Chiappella A, Calleri A, Fiori S, Tabanelli V, Cabras A, Pruneri G, Vitolo U, Gianni AM, Rambaldi A, Corradini P, Zinzani PL, Tarella C, Pileri S.
Subjects
NFKBIA, Diffuse Large B-cell Lymphoma, BCL-2, MYC, Gene expression profiling
Abstract
Recent randomized trials focused on gene expression-based determination of the cell of origin in diffuse large B-cell lymphoma could not show significant improvements by adding novel agents to standard chemoimmunotherapy. The aim of this study was the identification of a gene signature able to refine current prognostication algorithms and applicable to clinical practice. Here we used a targeted gene expression profiling panel combining the Lymph2Cx signature for cell of origin classification with additional targets including MYC, BCL-2 and NFKBIA, in 186 patients from 2 randomized trials (discovery cohort) (NCT00355199 and NCT00499018). Data were validated in 3 independent series (2 large public datasets and a real-life cohort). By integrating the cell of origin, MYC/BCL-2 double expressor status and NFKBIA expression, we defined a 3-gene signature combining MYC, BCL-2 and NFKBIA (MBN-signature), which outperformed the MYC/BCL-2 double expressor status in multivariate analysis, and allowed further risk stratification within the germinal center B-cell/unclassified subset. The high-risk (MBN Sig-high) subgroup identified the vast majority of double hit cases and a significant fraction of Activated B-Cell-derived diffuse large B-cell lymphomas. These results were validated in 3 independent series including a cohort from the REMoDL-B trial, where, in an exploratory ad hoc analysis, the addition of bortezomib in the MBN Sig-high subgroup provided a progression free survival advantage compared with standard chemoimmunotherapy. These data indicate that a simple 3-gene signature based on MYC, BCL-2 and NFKBIA could refine the prognostic stratification in diffuse large B-cell lymphoma, and might be the basis for future precision-therapy approaches.
Ciavarella S., Vegliante M. C., Fabbri M., De Summa S., Melle F., Motta G., De Iuliis V., Opinto G., Enjuanes A., Rega S., Gulino A., Agostinelli C., Scattone A., Tommasi S., Mangia A., Mele F., Simone G., Zito A. F., Ingravallo G., Vitolo U., Chiappella A., Tarella C., Gianni A. M., Rambaldi A., Zinzani P. L., Casadei B., Derenzini E., Loseto G., Pileri A., Tabanelli V., Fiori S., Rivas-Delgado A., Lopez-Guillermo A., Venesio T., Sapino A., Campo E., Tripodo C., Guarini A., Pileri S. A., Ciavarella S., Vegliante M.C., Fabbri M., De Summa S., Melle F., Motta G., De Iuliis V., Opinto G., Enjuanes A., Rega S., Gulino A., Agostinelli C., Scattone A., Tommasi S., Mangia A., Mele F., Simone G., Zito A.F., Ingravallo G., Vitolo U., Chiappella A., Tarella C., Gianni A.M., Rambaldi A., Zinzani P.L., Casadei B., Derenzini E., Loseto G., Pileri A., Tabanelli V., Fiori S., Rivas-Delgado A., Lopez-Guillermo A., Venesio T., Sapino A., Campo E., Tripodo C., Guarini A., and Pileri S.A.
Subjects
DLBCL microenvironment, gene expression DLBCL
Abstract
The affiliations of authors T. Venesio and A. Sapino were originally incorrect. These have now been corrected as per the author listing above.
Laginestra, M A, primary, Piccaluga, P P, additional, Fuligni, F, additional, Rossi, M, additional, Agostinelli, C, additional, Righi, S, additional, Sapienza, M R, additional, Motta, G, additional, Gazzola, A, additional, Mannu, C, additional, Sabattini, E, additional, Bacci, F, additional, Tabanelli, V, additional, Sacchetti, C A S, additional, Barrese, T Z, additional, Etebari, M, additional, Melle, F, additional, Clò, A, additional, Gibellini, D, additional, Tripodo, C, additional, Inghirami, G, additional, Croce, C M, additional, and Pileri, S A, additional
Sabattini, E., primary, Pizzi, M., additional, Tabanelli, V., additional, Baldin, P., additional, Sacchetti, C. S., additional, Agostinelli, C., additional, Zinzani, P. L., additional, and Pileri, S. A., additional
Laura Ballotta, Pier Luigi Zinzani, Stefano Pileri, Riccardo Bruna, Monica Tani, Beatrice Casadei, Valentina Tabanelli, Stefano Volpetti, Stefano Luminari, Paolo Corradini, Elisa Lucchini, Maria Chiara Tisi, Michele Merli, Alessandro Re, Marzia Varettoni, Emanuela Anna Pesce, Francesco Zaja, Ballotta, L., Zinzani, P. L., Pileri, S., Bruna, R., Tani, M., Casadei, B., Tabanelli, V., Volpetti, S., Luminari, S., Corradini, P., Lucchini, E., Tisi, M. C., Merli, M., Re, A., Varettoni, M., Pesce, E. A., Zaja, F., Ballotta L., Zinzani P.L., Pileri S., Bruna R., Tani M., Casadei B., Tabanelli V., Volpetti S., Luminari S., Corradini P., Lucchini E., Tisi M.C., Merli M., Re A., Varettoni M., Pesce E.A., and Zaja F.
Subjects
BCL2 inhibition, BCL2 protein, peripheral T-cell lymphoma, relapsed/refractory, venetoclax, Cancer Research, Oncology, Neoplasms. Tumors. Oncology. Including cancer and carcinogens, Clinical Trial, RC254-282
Abstract
Patients with relapsed/refractory (R/R) peripheral T-cell lymphoma (PTCL) have a poor prognosis, with an expected survival of less than 1 year using standard salvage therapies. Recent advances in our understanding of the biology of PTCL have led to identifying B-Cell Lymphoma 2 (BCL2) protein as a potential therapeutic target. BLC2 inhibitor venetoclax was investigated in a prospective phase II trial in patients with BCL2-positive R/R PTCL after at least one previous standard line of treatment (NCT03552692). Venetoclax given alone at a dosage of 800 mg/day resulted in one complete response (CR) and two stable diseases (SDs) among 17 enrolled patients. The majority of patients (88.2%) interrupted the treatment due to disease progression. No relationship with BCL2 expression was documented. At a median follow-up of 8 months, two patients are currently still on treatment (one CR and one SD). No case of tumor lysis syndrome was registered. Therefore, venetoclax monotherapy shows activity in a minority of patients whose biological characteristics have not yet been identified.Clinical Trial Registrationwww.clinicaltrials.gov (NCT03552692, EudraCT number 2017-004630-29).
Blastic Plasmacytoid Dendritic Cell Neoplasm is a rare and aggressive hematological malignancy currently lacking an effective therapy. To possibly identify genetic alterations useful for a new treatment design, we analyzed by whole-exome sequencing fourteen Blastic Plasmacytoid Dendritic Cell Neoplasm patients and the patient-derived CAL-1 cell line. The functional enrichment analysis of mutational data reported the epigenetic regulatory program as the most significantly undermined (P
Lorenzo Cerroni, Federica Melle, Marcello Del Corvo, Marco Paulli, Emilio Berti, Jessica Consiglio, Gaetano Ivan Dellino, Giovanna Motta, Stefano Pileri, Carlo M. Croce, Vincenzo Mazzara, Stefano Fiori, Fabio Fuligni, Giuseppe Benvenuto, Alessandro Pileri, Fabio Facchetti, Claudio Tripodo, Daniele Fanoni, Maria Rosaria Sapienza, Manuela Ferracin, Elena Sabattini, Valentina Tabanelli, Saveria Mazzara, Beatrice Belmonte, Sapienza M.R., Benvenuto G., Ferracin M., Mazzara S., Fuligni F., Tripodo C., Belmonte B., Fanoni D., Melle F., Motta G., Tabanelli V., Consiglio J., Mazzara V., Corvo M.D., Fiori S., Pileri A., Dellino G.I., Cerroni L., Facchetti F., Berti E., Sabattini E., Paulli M., Croce C.M., and Pileri S.A.
Subjects
Cancer Research, Neurogenesis, Neoplasms. Tumors. Oncology. Including cancer and carcinogens, MicroRNA Expression Profile, sequencing, Biology, Settore MED/08 - Anatomia Patologica, BPDCN, MiRNA, Network, Neurogenesis, Sequencing, BPDCN, Article, Chromatin, Gene expression profiling, MiRNA, Network, Sequencing, neurogenesis, Oncology, Downregulation and upregulation, microRNA, network, Cancer research, Immunohistochemistry, Settore MED/05 - Patologia Clinica, Neurogenesi, RC254-282, Progenitor, miRNA
Abstract
Simple Summary For the first time, neuronal features are described in blastic plasmacytoid dendritic cell neoplasm (BPDCN) by a complex array of molecular techniques, including microRNA and gene expression profiling, RNA and Chromatin immunoprecipitation sequencing, and immunohistochemistry. The discovery of unexpected neural features in BPDCN may change our vision of this disease, leading to the designing of a new BPDCN cell model and to re-thinking the relations occurring between BPDCN and nervous system. The observed findings contribute to explaining the extreme tumor aggressiveness and also to propose novel therapeutic targets. In view of this, the identification, in this work of new potential neural metastatic inducers might open the way to therapeutic approaches for BPDCN patients based on the use of anti-neurogenic agents. Abstract Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and highly aggressive hematologic malignancy originating from plasmacytoid dendritic cells (pDCs). The microRNA expression profile of BPDCN was compared to that of normal pDCs and the impact of miRNA dysregulation on the BPDCN transcriptional program was assessed. MiRNA and gene expression profiling data were integrated to obtain the BPDCN miRNA-regulatory network. The biological process mainly dysregulated by this network was predicted to be neurogenesis, a phenomenon raising growing interest in solid tumors. Neurogenesis was explored in BPDCN by querying different molecular sources (RNA sequencing, Chromatin immunoprecipitation-sequencing, and immunohistochemistry). It was shown that BPDCN cells upregulated neural mitogen genes possibly critical for tumor dissemination, expressed neuronal progenitor markers involved in cell migration, exchanged acetylcholine neurotransmitter, and overexpressed multiple neural receptors that may stimulate tumor proliferation, migration and cross-talk with the nervous system. Most neural genes upregulated in BPDCN are currently investigated as therapeutic targets.
0301 basic medicine, Oncology, medicine.medical_specialty, diagnosis, diffuse large B-cell lymphoma, Review, Settore MED/08 - Anatomia Patologica, 03 medical and health sciences, 0302 clinical medicine, Internal medicine, Daily practice, medicine, Tumor Microenvironment, Humans, lcsh:QH301-705.5, B cell, therapy, business.industry, Gene Expression Profiling, Not Otherwise Specified, High-Throughput Nucleotide Sequencing, General Medicine, medicine.disease, Microarray Analysis, Prognosis, Lymphoma, Gene expression profiling, diagnosi, 030104 developmental biology, medicine.anatomical_structure, lcsh:Biology (General), Lymphoid malignancy, classification, 030220 oncology & carcinogenesis, next-generation sequencing, Lymphoma, Large B-Cell, Diffuse, business, Diffuse large B-cell lymphoma, prognosi
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the commonest form of lymphoid malignancy, with a prevalence of about 40% worldwide. Its classification encompasses a common form, also termed as “not otherwise specified” (NOS), and a series of variants, which are rare and at least in part related to viral agents. Over the last two decades, DLBCL-NOS, which accounts for more than 80% of the neoplasms included in the DLBCL chapter, has been the object of an increasing number of molecular studies which have led to the identification of prognostic/predictive factors that are increasingly entering daily practice. In this review, the main achievements obtained by gene expression profiling (with respect to both neoplastic cells and the microenvironment) and next-generation sequencing will be discussed and compared. Only the amalgamation of molecular attributes will lead to the achievement of the long-term goal of using tailored therapies and possibly chemotherapy-free protocols capable of curing most (if not all) patients with minimal or no toxic effects.
Stefano Pileri, Anna Rotili, Claudio Agostinelli, Chiara Corsini, Valentina Tabanelli, Federica Melle, Arianna Di Napoli, Angelica Calleri, Stefania Orecchioni, Giovanna Motta, Stefano Fiori, Tabanelli V., Corsini C., Fiori S., Agostinelli C., Calleri A., Orecchioni S., Melle F., Motta G., Rotili A., Di Napoli A., and Pileri S.A.
Subjects
0301 basic medicine, Adult, STAT3 Transcription Factor, Anaplastic Lymphoma, DNA Copy Number Variations, Breast Implants, Breast Neoplasms, In situ hybridization, B7-H1 Antigen, Pathology and Forensic Medicine, 03 medical and health sciences, 0302 clinical medicine, hemic and lymphatic diseases, Breast implant, PDL1, Gene duplication, medicine, Biomarkers, Tumor, Humans, Phosphorylation, STAT3, Anaplastic large-cell lymphoma, Aged, Polysomy, anaplastic lymphoma, breast implant, copy gain, pdl1, polysomy, biology, Copy gain, JAK-STAT signaling pathway, Middle Aged, medicine.disease, Immunohistochemistry, 030104 developmental biology, 030220 oncology & carcinogenesis, Anaplastic lymphoma, biology.protein, Cancer research, Lymphoma, Large-Cell, Anaplastic, Female
Abstract
Breast implant-associated anaplastic large cell lymphoma (BI-ALCL) is a variant of anaplastic large cell lymphoma arising within seroma effusion associated with breast implants. BI-ALCL is a rare disease, recently recognized as a new provisional entity by the 2017 revised World Health Organization classification. All BI-ALCLs tested so far showed a "triple-negative" genetic profile-negative for ALK, DUSP22, and TP63 rearrangements-and were characterized by mutational and gene expression profiles consistent with aberrant activation of the JAK/STAT pathway. The active form of STAT3 (pSTAT3) is constantly expressed in BI-ALCLs and may favor tumor immune escape by triggering the transcription of PDL1 (CD274), a gene encoding the immune-checkpoint molecule programmed cell death ligand 1 (PDL1); immunohistochemical positivity for PDL1 has been recently described in 3 BI-ALCL cases, and one of them also harbored PDL1 gene amplification. We evaluated PDL1 and pSTAT expression by immunohistochemistry and PDL1 copy number alterations (CNAs) at chromosome 9p24.1 by fluorescent in situ hybridization in a cohort of 9 BI-ALCL cases; we also investigated the presence of tumor-infiltrating programmed cell death 1 (PD1)+ T cells (tumor-infiltrating lymphocytes, or TILs) and PDL1+ tumor-associated macrophages (TAMs) in BI-ALCL microenvironment. Tumor cells expressed PDL1 in 5 (56%) of 9 cases and harbored PDL1 CNAs in 3 (33%) of 9 cases; immunohistochemistry for pSTAT3 was positive in all 6 cases tested (100%), indicative of active JAK/STAT signaling. We observed PDL1 CNAs only among PDL1-positive cases, whereas PD1+ TILs and PDL1+ TAMs were present at variable levels in both PDL1-positive and PDL1-negative BI-ALCLs. We report frequent PDL1 expression and recurrent PDL1 CNAs in BI-ALCLs: our data suggest that 9p24.1 alterations represent a common mechanism of PDL1 overexpression in this disease, likely acting in synergy with constitutive pSTAT3 signaling. In PDL1-positive cases without chromosomal aberration, PDL1 expression may be induced by JAK/STAT signaling alone and/or others alternative pathways. BI-ALCL microenvironment hosts variable amounts of PD1+ TILs and PDL1+ TAMs, suggesting the presence of an active PD1/PDL1 axis. These findings may be of therapeutic value in advanced-stage patients who may benefit from a PD1/PDL1 blocking treatment.
Claudio Agostinelli, Federica Melle, Giorgio Inghirami, Tabanelli, Anna Gazzola, Martina Rossi, G Motta, Alberto Clò, Simona Righi, Fabio Fuligni, Tomas Barrese, Pier Paolo Piccaluga, Claudio Tripodo, Carlo Alberto Sagramoso Sacchetti, Maria Antonella Laginestra, Davide Gibellini, Stefano Pileri, Claudia Mannu, Elena Sabattini, Francesco Bacci, Carlo M. Croce, Maryam Etebari, Maria Rosaria Sapienza, Laginestra, M., Piccaluga, P., Fuligni, F., Rossi, M., Agostinelli, C., Righi, S., Sapienza, M., Motta, G., Gazzola, A., Mannu, C., Sabattini, E., Bacci, E., Tabanelli, V., Sacchetti, C., Barrese, T., Etebari, M., Melle, F., Clò, A., Gibellini, D., Tripodo, C., Inghirami, G., Croce, C., Pileri, S., Laginestra, M.A, Piccaluga, P.P., Sapienza, M.R., Sagramoso Sacchetti, C.A., Barrese, T.Z., Croce, C.M., and Pileri, S.A.
Subjects
Female, Gene Expression Profiling, Humans, Lymphoma, T-Cell, Peripheral, Male, MicroRNAs, Oligonucleotide Array Sequence Analysis, RNA, Neoplasm, Gene Expression Regulation, Neoplastic, Oncology, Hematology, Medicine (all), medicine.medical_specialty, Pathology, Peripheral T-cell lymphoma not otherwise specified, Biology, hemic and lymphatic diseases, Internal medicine, microRNA, medicine, Regulation of gene expression, PTCLs/NOS, GEP, Oligonucleotide Array Sequence Analysi, Not Otherwise Specified, MicroRNA, medicine.disease, Lymphoma, Gene expression profiling, Original Article, CD8, Human
Abstract
Peripheral T-cell lymphomas not otherwise specified (PTCLs/NOS) are rare and aggressive tumours whose molecular pathogenesis and diagnosis are still challenging. The microRNA (miRNA) profile of 23 PTCLs/NOS was generated and compared with that of normal T-lymphocytes (CD4+, CD8+, naive, activated). The differentially expressed miRNA signature was compared with the gene expression profile (GEP) of the same neoplasms. The obtained gene patterns were tested in an independent cohort of PTCLs/NOS. The miRNA profile of PTCLs/NOS then was compared with that of 10 angioimmunoblastic T-cell lymphomas (AITLs), 6 anaplastic large-cell lymphomas (ALCLs)/ALK+ and 6 ALCLs/ALK - . Differentially expressed miRNAs were validated in an independent set of 20 PTCLs/NOS, 20 AITLs, 19 ALCLs/ALK - and 15 ALCLs/ALK+. Two hundred and thirty-six miRNAs were found to differentiate PTCLs/NOS from activated T-lymphocytes. To assess which miRNAs impacted on GEP, a multistep analysis was performed, which identified all miRNAs inversely correlated to different potential target genes. One of the most discriminant miRNAs was selected and its expression was found to affect the global GEP of the tumours. Moreover, two sets of miRNAs were identified distinguishing PTCL/NOS from AITL and ALCL/ALK - , respectively. The diagnostic accuracy of this tool was very high (83.54%) and its prognostic value validated.
Claudio Agostinelli, Stefano Pileri, Valentina Tabanelli, Simona Righi, Claudia Mannu, Maria Teresa Sista, Saveria Capria, Anna Gazzola, Pier Paolo Piccaluga, Elena Sabattini, Giovanna Meloni, Francesco Bacci, Tabanelli V., Agostinelli C., Sabattini E., Gazzola A., Bacci F., Capria S., Mannu C., Righi S., Sista M.T., Meloni G., Pileri S.A., and Piccaluga P.P.
Subjects
Epstein-Barr-viru, Pathology, medicine.medical_specialty, PTCL, Fulminant, T cell, Hepatosplenomegaly, Lymphoproliferative disorders, lcsh:Medicine, Case Report, Disease, EBV, hemic and lymphatic diseases, medicine, Cytotoxic T cell, Medicine(all), business.industry, lcsh:R, Peripheral T-cell lymphoma, General Medicine, medicine.disease, Pancytopenia, medicine.anatomical_structure, medicine.symptom, business, CD8
Abstract
Introduction Systemic Epstein-Barr-virus-positive T cell lymphoproliferative disease of childhood is an extremely rare disorder, characterized by clonal proliferation of Epstein-Barr-virus-infected T cells with an activated cytotoxic phenotype. The disease is more frequent in Asia and South America, with only few cases reported in Western countries. A prompt diagnosis, though often difficult, is a necessity due to the very aggressive clinical course of the disease. Case presentation We report the clinicopathological features of fulminant T cell lymphoproliferative disease that arose in the setting of acute primary Epstein-Barr virus infection. Our patient, a 23-year-old man, presented to our facility with persisting fever, hepatosplenomegaly and severe pancytopenia. On bone marrow biopsy, an abundant lymphoid infiltrate was observed. Immunophenotypic and molecular studies revealed that the atypical lymphoid cells displayed a CD8+, Epstein-Barr-encoded-RNA-positive T cell phenotype with clonal rearrangement of the T cell receptor genes, the final diagnosis being systemic Epstein-Barr-virus-positive T cell lymphoproliferative disease. On reviewing the literature we found only 14 similar cases, all presenting with very aggressive clinical courses and requiring extensive phenotyping and molecular techniques for final diagnosis. Conclusion Though extremely rare, this disease can occur in Europe, and a comprehensive diagnostic approach is thus recommended in all case of Epstein-Barr-virus-positive lymphoproliferative disorders. Unfortunately, at present no specific treatment is available; however, prompt administration of anti- Epstein-Barr virus treatment and rapid attempts to control the hemophagocytic syndrome are indicated.
Gargano G, Vegliante MC, Esposito F, Pappagallo SA, Sabattini E, Agostinelli C, Pileri SA, Tabanelli V, Ponzoni M, Lorenzi L, Facchetti F, Di Napoli A, Lucioni M, Paulli M, Leoncini L, Lazzi S, Ascani S, Opinto G, Zaccaria GM, Volpe G, Mondelli P, Bucci A, Selicato L, Negri A, Loseto G, Clemente F, Scattone A, Zito AF, Nassi L, Del Buono N, Guarini A, and Ciavarella S
Tedeschi A, Frustaci AM, Condoluci A, Coscia M, Chiarle R, Zinzani PL, Motta M, Gaidano G, Quaresmini G, Scarfò L, Catania G, Deodato M, Jones R, Tabanelli V, Griggio V, Stüssi G, Calleri A, Pini K, Cairoli R, Zenz T, Signori A, Zucca E, Rossi D, and Montillo M
Sangiorgio V, Palasciano A, Tabanelli V, Giné E, Guerra L, Pagni F, Casiraghi A, Casaroli I, Frigola G, Magnano L, Gambacorti-Passerini C, Derenzini E, Vanazzi A, and Campo E
Mark E, Kempf W, Guitart J, Pulitzer M, Mitteldorf C, Hristov A, Torres-Cabala C, Marchi E, Cropley T, Rodriguez Pinilla SM, Griffin T, Fernandez R, Pileri S, Pileri A, Tabanelli V, Borretta L, Subtil A, Plaza JA, Piris JAMA, Feldman AL, Cerroni L, and Gru AA
Arcari A, Tabanelli V, Merli F, Marcheselli L, Merli M, Balzarotti M, Zilioli VR, Fabbri A, Cavallo F, Casaluci GM, Tucci A, Puccini B, Pennese E, Di Rocco A, Zanni M, Flenghi L, Gini G, Sartori R, Chiappella A, Usai SV, Tani M, Marino D, Arcaini L, Vallisa D, and Spina M
Subjects
Aged, Humans, Hepacivirus genetics, Antiviral Agents therapeutic use, Prospective Studies, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasm Recurrence, Local drug therapy, Rituximab therapeutic use, Doxorubicin therapeutic use, Vincristine therapeutic use, Cyclophosphamide therapeutic use, Prednisone therapeutic use, Hepatitis C, Chronic drug therapy, Hepatitis C drug therapy, Hepatitis C epidemiology, Lymphoma, Large B-Cell, Diffuse
Purpose: Peripheral T-cell lymphoma (PTCL) includes heterogeneous clinicopathologic entities with numerous diagnostic and treatment challenges. We previously defined robust transcriptomic signatures that distinguish common PTCL entities and identified two novel biologic and prognostic PTCL-not otherwise specified subtypes (PTCL-TBX21 and PTCL-GATA3). We aimed to consolidate a gene expression-based subclassification using formalin-fixed, paraffin-embedded (FFPE) tissues to improve the accuracy and precision in PTCL diagnosis., Materials and Methods: We assembled a well-characterized PTCL training cohort (n = 105) with gene expression profiling data to derive a diagnostic signature using fresh-frozen tissue on the HG-U133plus2.0 platform (Affymetrix, Inc, Santa Clara, CA) subsequently validated using matched FFPE tissues in a digital gene expression profiling platform (nCounter, NanoString Technologies, Inc, Seattle, WA). Statistical filtering approaches were applied to refine the transcriptomic signatures and then validated in another PTCL cohort (n = 140) with rigorous pathology review and ancillary assays., Results: In the training cohort, the refined transcriptomic classifier in FFPE tissues showed high sensitivity (> 80%), specificity (> 95%), and accuracy (> 94%) for PTCL subclassification compared with the fresh-frozen-derived diagnostic model and showed high reproducibility between three independent laboratories. In the validation cohort, the transcriptional classifier matched the pathology diagnosis rendered by three expert hematopathologists in 85% (n = 119) of the cases, showed borderline association with the molecular signatures in 6% (n = 8), and disagreed in 8% (n = 11). The classifier improved the pathology diagnosis in two cases, validated by clinical findings. Of the 11 cases with disagreements, four had a molecular classification that may provide an improvement over pathology diagnosis on the basis of overall transcriptomic and morphological features. The molecular subclassification provided a comprehensive molecular characterization of PTCL subtypes, including viral etiologic factors and translocation partners., Conclusion: We developed a novel transcriptomic approach for PTCL subclassification that facilitates translation into clinical practice with higher precision and uniformity than conventional pathology diagnosis.
Roma S, Camisaschi C, Mancuso P, Trabanelli S, Vanazzi A, Villa S, Prati D, Fiori S, Lorenzini D, Tabanelli V, Pileri S, Tarella C, Jandus C, and Bertolini F
Vegliante MC, Mazzara S, Zaccaria GM, De Summa S, Esposito F, Melle F, Motta G, Sapienza MR, Opinto G, Volpe G, Bucci A, Gargano G, Enjuanes A, Tabanelli V, Fiori S, Minoia C, Clemente F, Negri A, Gulino A, Morello G, Scattone A, Zito AF, Tommasi S, Agostinelli C, Vitolo U, Chiappella A, Barbui AM, Derenzini E, Zinzani PL, Casadei B, Rivas-Delgado A, López-Guillermo A, Campo E, Moschetta A, Guarini A, Pileri SA, and Ciavarella S
Subjects
Humans, Tumor Microenvironment, Liver X Receptors genetics, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics
Tabanelli V, Melle F, Motta G, Mazzara S, Fabbri M, Agostinelli C, Calleri A, Del Corvo M, Fiori S, Lorenzini D, Cesano A, Chiappella A, Vitolo U, Derenzini E, Griffin GK, Rodig SJ, Vanazzi A, Sabattini E, Tarella C, Sapienza MR, and Pileri SA
Subjects
Apoptosis, Hepatocyte Nuclear Factor 1-alpha, Histiocytes metabolism, Histiocytes pathology, Humans, Ligands, Programmed Cell Death 1 Receptor, Tumor Microenvironment, B7-H1 Antigen genetics, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics
Background: The mechanisms involved in mycosis fungoides, and Sezary Syndrome progression are largely unknown. Over the last decade the interest in immune system contrast of neoplasm has grown owing to the introduction of immunotherapy. PD-1 and its ligand (PD-L1) are the target of several immunotherapy treatment. In the literature reports on the expression of PD-1 and PD-L1 have provided contrasting results., Methods: In our analysis we investigated PD-1 expression in neoplastic cells and in tumor infiltrating lymphocytes (TILs) as well as PD-L1 expression in tumor cells and in tumor associated macrophages (TAMs). PD-L1 and PD-1 positive cells were counted in 5 high-power fields (HPF) and scored as the average number of positive neoplastic cells/TILs/TAMs per HPF., Results: From databases of two institutions (Bologna and Florence) thirty-five patients corresponding to 43 biopsies were retrieved. In seven instances sequential biopsies were present. No statistically significant expression was observed comparing early to advanced stages by analysing PD-1 by tumor cells and TILs and of PD-L1 by tumor cells and TAMs., Conclusions: Our results corroborate that PD-1 and PD-L1 expression is not stage-dependent in mycosis fungoides and Sezary syndrome. However, PD-1 and PD-L1 expression in affected patients provides a rationale to schedule anti PD-1/PD-L1 drugs.
Zaccaria GM, Colella V, Colucci S, Clemente F, Pavone F, Vegliante MC, Esposito F, Opinto G, Scattone A, Loseto G, Minoia C, Rossini B, Quinto AM, Angiulli V, Grieco LA, Fama A, Ferrero S, Moia R, Di Rocco A, Quaglia FM, Tabanelli V, Guarini A, and Ciavarella S
Subjects
Disease Management, Humans, Natural Language Processing, Workflow, Electronic Health Records, Hematology methods, Hematology standards, Medical Oncology methods, Medical Oncology standards, Research Report
Ballotta L, Zinzani PL, Pileri S, Bruna R, Tani M, Casadei B, Tabanelli V, Volpetti S, Luminari S, Corradini P, Lucchini E, Tisi MC, Merli M, Re A, Varettoni M, Pesce EA, and Zaja F
Sapienza MR, Benvenuto G, Ferracin M, Mazzara S, Fuligni F, Tripodo C, Belmonte B, Fanoni D, Melle F, Motta G, Tabanelli V, Consiglio J, Mazzara V, Del Corvo M, Fiori S, Pileri A, Dellino GI, Cerroni L, Facchetti F, Berti E, Sabattini E, Paulli M, Croce CM, and Pileri SA
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and highly aggressive hematologic malignancy originating from plasmacytoid dendritic cells (pDCs). The microRNA expression profile of BPDCN was compared to that of normal pDCs and the impact of miRNA dysregulation on the BPDCN transcriptional program was assessed. MiRNA and gene expression profiling data were integrated to obtain the BPDCN miRNA-regulatory network. The biological process mainly dysregulated by this network was predicted to be neurogenesis, a phenomenon raising growing interest in solid tumors. Neurogenesis was explored in BPDCN by querying different molecular sources (RNA sequencing, Chromatin immunoprecipitation-sequencing, and immunohistochemistry). It was shown that BPDCN cells upregulated neural mitogen genes possibly critical for tumor dissemination, expressed neuronal progenitor markers involved in cell migration, exchanged acetylcholine neurotransmitter, and overexpressed multiple neural receptors that may stimulate tumor proliferation, migration and cross-talk with the nervous system. Most neural genes upregulated in BPDCN are currently investigated as therapeutic targets.
Pileri SA, Tabanelli V, Fiori S, Calleri A, Melle F, Motta G, Lorenzini D, Tarella C, and Derenzini E
Abstract
Peripheral T-cell lymphoma, not otherwise specified (PTCL_NOS) corresponds to about one fourth of mature T-cell tumors, which overall represent 10-12% of all lymphoid malignancies. This category comprises all T-cell neoplasms, which do not correspond to any of the distinct entities listed in the WHO (World Health Organization) Classification of Tumours of Haematopoietic and Lymphoid Tissues. In spite of the extreme variability of morphologic features and phenotypic profiles, gene expression profiling (GEP) studies have shown a signature that is distinct from that of all remaining PTCLs. GEP has also allowed the identification of subtypes provided with prognostic relevance. Conversely to GEP, next-generation sequencing (NGS) has so far been applied to a limited number of cases, providing some hints to better understand the pathobiology of PTCL_NOS. Although several pieces of information have emerged from pathological studies, PTCL_NOS still remains a tumor with a dismal prognosis. The usage of CHOEP (cyclophosphamide, doxorubicin, vincristine, prednisone, etoposide) followed by autologous stem cell transplantation may represent the best option, by curing about 50% of the patients whom such an approach can be applied to. Many new drugs have been proposed without achieving the expected results. Thus, the optimal treatment of PTCL_NOS remains unidentified.
Derenzini E, Mazzara S, Melle F, Motta G, Fabbri M, Bruna R, Agostinelli C, Cesano A, Corsini CA, Chen N, Righi S, Sabattini E, Chiappella A, Calleri A, Fiori S, Tabanelli V, Cabras A, Pruneri G, Vitolo U, Gianni AM, Rambaldi A, Corradini P, Zinzani PL, Tarella C, and Pileri S
Recent randomized trials focused on gene expression-based determination of the cell of origin in diffuse large B-cell lymphoma could not show significant improvements by adding novel agents to standard chemoimmunotherapy. The aim of this study was the identification of a gene signature able to refine current prognostication algorithms and applicable to clinical practice. Here we used a targeted gene expression profiling panel combining the Lymph2Cx signature for cell of origin classification with additional targets including MYC, BCL-2 and NFKBIA, in 186 patients from 2 randomized trials (discovery cohort) (NCT00355199 and NCT00499018). Data were validated in 3 independent series (2 large public datasets and a real-life cohort). By integrating the cell of origin, MYC/BCL-2 double expressor status and NFKBIA expression, we defined a 3-gene signature combining MYC, BCL-2 and NFKBIA (MBN-signature), which outperformed the MYC/BCL-2 double expressor status in multivariate analysis, and allowed further risk stratification within the germinal center B-cell/unclassified subset. The high-risk (MBN Sig-high) subgroup identified the vast majority of double hit cases and a significant fraction of Activated B-Cell-derived diffuse large B-cell lymphomas. These results were validated in 3 independent series including a cohort from the REMoDL-B trial, where, in an exploratory ad hoc analysis, the addition of bortezomib in the MBN Sig-high subgroup provided a progression free survival advantage compared with standard chemoimmunotherapy. These data indicate that a simple 3-gene signature based on MYC, BCL-2 and NFKBIA could refine the prognostic stratification in diffuse large B-cell lymphoma, and might be the basis for future precision-therapy approaches.
Rotili A, Ferrari F, Nicosia L, Pesapane F, Tabanelli V, Fiori S, Vanazzi A, Meneghetti L, Abbate F, Latronico A, and Cassano E
Subjects
Breast diagnostic imaging, Female, Humans, Breast Implants adverse effects, Breast Neoplasms diagnostic imaging, Breast Neoplasms etiology, Lymphoma, Large-Cell, Anaplastic diagnostic imaging, Lymphoma, Large-Cell, Anaplastic etiology, Magnetic Resonance Imaging methods
Abstract
Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) is a rare and newly recognized subtype of T cell Non-Hodgkin Lymphoma (NHLs) associated with breast implants.The mechanism involved in the development of this kind of lymphoma is still uncertain.BIA-ALCL is generally an indolent disease localized to the breast implant and its capsule and effectively treated with capsulectomy alone without chemotherapy.Clinically, BIA-ALCL may typically present a sudden-onset breast-swelling secondary to periimplant effusion. The minority of BIA-ALCL patients present a more aggressive mass-forming subtype, for which systemic therapy is mandatory.Despite the number of cases has recently increased, BIA-ALCL remains a rare disease described mainly in several case reports and small case series.Breast imaging, including mammography, ultrasound and breast MRI are routinely used in the screening of breast cancer; however, guidelines for the imaging and pathological diagnosis of this disease have only recently been proposed and included in the 2019 National Comprehensive Cancer Network (NCCN) consensus guidelines for BIA-ALCL.The main purpose of this pictorial is to illustrate the MRI signs of BIA-ALCL and correlate them with the corresponding pathology features in order to improve the knowledge of the principals MRI features of this type of lymphoma.
Diffuse large B-cell lymphoma (DLBCL) is the commonest form of lymphoid malignancy, with a prevalence of about 40% worldwide. Its classification encompasses a common form, also termed as "not otherwise specified" (NOS), and a series of variants, which are rare and at least in part related to viral agents. Over the last two decades, DLBCL-NOS, which accounts for more than 80% of the neoplasms included in the DLBCL chapter, has been the object of an increasing number of molecular studies which have led to the identification of prognostic/predictive factors that are increasingly entering daily practice. In this review, the main achievements obtained by gene expression profiling (with respect to both neoplastic cells and the microenvironment) and next-generation sequencing will be discussed and compared. Only the amalgamation of molecular attributes will lead to the achievement of the long-term goal of using tailored therapies and possibly chemotherapy-free protocols capable of curing most (if not all) patients with minimal or no toxic effects.
Sapienza MR, Fuligni F, Melle F, Tabanelli V, Indio V, Laginestra MA, Motta G, Mazzara S, Cerroni L, Pileri A, Facchetti F, Paulli M, Cascione L, Laganà A, Berti E, Ferracin M, Agostinelli C, Sabattini E, Croce CM, and Pileri SA
Peripheral T-cell lymphoma not otherwise specified represents a diagnostic category comprising clinically, histologically, and molecularly heterogeneous neoplasms that are poorly understood. The genetic landscape of peripheral T-cell lymphoma not otherwise specified remains largely undefined, only a few sequencing studies having been conducted so far. In order to improve our understanding of the genetics of this neoplasm, we performed whole exome sequencing along with RNA-sequencing in a discovery set of 21 cases. According to whole exome sequencing results and mutations previously reported in other peripheral T-cell lymphomas, 137 genes were sequenced by a targeted deep approach in 71 tumor samples. In addition to epigenetic modifiers implicated in all subtypes of T-cell neoplasm (TET2, DNMT3A, KMT2D, KMT2C, SETD2), recurrent mutations of the FAT1 tumor suppressor gene were for the first time recorded in 39% of cases. Mutations of the tumor suppressor genes LATS1, STK3, ATM, TP53, and TP63 were also observed, although at a lower frequency. Patients with FAT1 mutations showed inferior overall survival compared to those with wild-type FAT1. Although peripheral T-cell lymphoma not otherwise specified remains a broad category also on molecular grounds, the present study highlights that FAT1 mutations occur in a significant proportion of cases, being provided with both pathogenetic and prognostic impact.
Sapienza MR, Pileri A, Derenzini E, Melle F, Motta G, Fiori S, Calleri A, Pimpinelli N, Tabanelli V, and Pileri S
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an extremely rare tumour, which usually affects elderly males and presents in the skin with frequent involvement of the bone-marrow, peripheral blood and lymph nodes. It has a dismal prognosis, with most patients dying within one year when treated by conventional chemotherapies. The diagnosis is challenging, since neoplastic cells can resemble lymphoblasts or small immunoblasts, and require the use of a large panel of antibodies, including those against CD4, CD56, CD123, CD303, TCL1, and TCF4. The morphologic and in part phenotypic ambiguity explains the uncertainties as to the histogenesis of the neoplasm that led to the use of various denominations. Recently, a series of molecular studies based on karyotyping, gene expression profiling, and next generation sequencing, have largely unveiled the pathobiology of the tumour and proposed the potentially beneficial use of new drugs. The latter include SL-401, anti-CD123 immunotherapies, venetoclax, BET-inhibitors, and demethylating agents. The epidemiologic, clinical, diagnostic, molecular, and therapeutic features of BPDCN are thoroughly revised in order to contribute to an up-to-date approach to this tumour that has remained an orphan disease for too long.
Pileri SA, Derenzini E, Melle F, Motta G, Calleri A, Antoniotti P, Maltoni V, Spagnolo S, Fiori S, Tabanelli V, and Fabbri M
Subjects
Animals, DNA Copy Number Variations, Humans, Sequence Analysis, DNA, Transcriptome, Lymphoma, Large B-Cell, Diffuse classification, Precision Medicine methods, Translational Research, Biomedical methods
Abstract
The updated edition of the Classification of Tumours of Haematopoietic and Lymphoid Tissues, published in September 2017 by the World Health Organization (WHO), presents many important changes to the document published in 2008. Most of these novelties are linked to the exceptional development of biomolecular techniques during the last 10 years. To illustrate how much new technologies have contributed to the better classification of single entities, as well as the discovery of new ones, would go beyond the objectives of this work. For this reason, we will take diffuse large B-cell lymphoma as an example of the cognitive improvement produced by high-yield technologies (such as the gene expression profile, the study of copy number variation, and the definition of the mutational spectrum). The acquisition of this knowledge not only has a speculative value but also represents the elements for effective application in daily practice. On the one hand, it would allow the development of personalised therapy programs, and on the other it would promote the transition from the bench of the researcher's laboratory to the patient's bedside., Competing Interests: No competing interests were disclosed.No competing interests were disclosed.No competing interests were disclosed.
De Luca G, Trasarti S, Bizzoni L, Del Giudice I, Della Starza I, De Propris MS, Gentile G, Mancini F, Mantovani S, Petrucci L, Tabanelli V, Guarini A, Vignetti M, and Foà R