23 results on '"Silvia Gimondi"'
Search Results
2. Data from Pharmacologic Inhibition of JAK1/JAK2 Signaling Reduces Experimental Murine Acute GVHD While Preserving GVT Effects
- Author
-
Jacopo Mariotti, Paolo Corradini, Anisa Bermema, Davide Confalonieri, Camilla Recordati, Antonio Vendramin, Silvia Gimondi, and Cristiana Carniti
- Abstract
Purpose: Immune-mediated graft-versus-tumor (GVT) effects can occur after allogeneic hematopoietic stem cell transplantation (HSCT), but GVT is tightly linked to its main complication, graft-versus-host disease (GVHD). Strategies aimed at modulating GVHD, while maintaining the GVT effect, are needed to improve the cure rate of transplant. Given the emerging role of Janus-activated kinase (JAK) signaling in lymphoproliferative and myeloproliferative diseases and its established function at dictating T-cell differentiation, we postulated that JAKs might be potential therapeutic targets through a pharmacologic approach.Experimental Design: We examined the effect of JAK1/JAK2 modulation by ruxolitinib in a mouse model of fully MHC mismatched bone marrow transplant comprising in vivo tumor inoculation.Results: JAK1/JAK2 inhibition by ruxolitinib improved both overall survival (P = 0.03) and acute GVHD pathologic score at target organs (P ≤ 0.001) of treated mice. In addition, treatment with ruxolitinib was associated with a preserved GVT effect, as evidenced by reduction of tumor burden (P = 0.001) and increase of survival time (P = 0.01). JAK1/JAK2 inhibition did not impair the in vivo acquisition of donor T-cell alloreactivity; this observation may account, at least in part, to the preserved GVT effect. Rather, JAK1/JAK2 inhibition of GVHD was associated with the modulation of chemokine receptor expression, which may have been one factor in the reduced infiltration of donor T cells in GVHD target organs.Conclusions: These data provide further evidence that JAK inhibition represents a new and potentially clinically relevant approach to GVHD prevention. Clin Cancer Res; 21(16); 3740–9. ©2015 AACR.
- Published
- 2023
3. Noninvasive Molecular Monitoring in Multiple Myeloma Patients Using Cell-Free Tumor DNA
- Author
-
Cristiana Carniti, Silvia Gimondi, Giulia Biancon, Paolo Corradini, and Antonio Vendramin
- Subjects
business.industry ,Context (language use) ,Gene rearrangement ,Cell free ,Ion semiconductor sequencing ,medicine.disease ,Minimal residual disease ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Molecular Medicine ,Bone marrow ,business ,Multiple myeloma ,030215 immunology - Abstract
Novel treatments for multiple myeloma (MM) have increased rates of complete response, raising interest in more accurate methods to evaluate residual disease. Cell-free tumor DNA (cfDNA) analysis may represent a minimally invasive approach complementary to multiparameter flow cytometry (MFC) and molecular methods on bone marrow aspirates. A sequencing approach using the Ion Torrent Personal Genome Machine was applied to identify clonal IGH gene rearrangements in tumor plasma cells (PCs) and in serial plasma samples of 25 patients with MM receiving second-line therapy. The same clonal IGH rearrangement identified in tumor PCs was detected in paired plasma samples, and levels of IGH cfDNA correlated with outcome and mirrored tumor dynamics evaluated using conventional laboratory parameters. In addition, IGH cfDNA levels reflected the number of PCs enumerated by MFC immunophenotyping even in the complete response context. Patients determined by MFC to be free of minimal residual disease were characterized by low frequencies of tumor clonotypes in cfDNA and longer survival. This pilot study supports the clinical applicability of the noninvasive monitoring of tumor levels in plasma samples of patients with MM by IGH sequencing.
- Published
- 2018
4. Hyperthermic Intraperitoneal Chemotherapy (HIPEC) at the Time of Primary Curative Surgery in Patients with Colorectal Cancer at High Risk for Metachronous Peritoneal Metastases
- Author
-
Ermanno Leo, Filippo Pietrantonio, Serena Bonomi, Marcello Guaglio, Dario Baratti, Shigeki Kusamura, Marcello Deraco, Domenico Rosario Iusco, Antonio Grassi, Massimo Milione, Silvia Gimondi, and Salvatore Virzì
- Subjects
Adult ,Male ,medicine.medical_specialty ,Colorectal cancer ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,medicine ,Humans ,Cumulative incidence ,Prospective Studies ,Prospective cohort study ,Peritoneal Neoplasms ,Aged ,business.industry ,Hazard ratio ,Cancer ,Neoplasms, Second Primary ,Common Terminology Criteria for Adverse Events ,Hyperthermia, Induced ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Surgery ,Oxaliplatin ,Italy ,Oncology ,030220 oncology & carcinogenesis ,Female ,030211 gastroenterology & hepatology ,Hyperthermic intraperitoneal chemotherapy ,Colorectal Neoplasms ,business ,medicine.drug - Abstract
Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) are maximally effective in early-stage colorectal cancer peritoneal metastases (CRC-PM); however, the use of HIPEC to treat subclinical-stage PM remains controversial. This prospective two-center study assessed adjuvant HIPEC in CRC patients at high risk for metachronous PM ( www.clinicaltrials.gov NCT02575859). During 2006–2012, a total of 22 patients without systemic metastases were prospectively enrolled to receive HIPEC simultaneously with curative surgery, plus adjuvant systemic chemotherapy (oxaliplatin/irinotecan-containing ± biologics), based on primary tumor-associated criteria: resected synchronous ovarian (n = 2) or minimal peritoneal (n = 6) metastases, primaries directly invading other organs (n = 4) or penetrating the visceral peritoneum (n = 10). A control group retrospectively included 44 matched (1:2) patients undergoing standard treatments and no HIPEC during the same period. The cumulative PM incidence was calculated in a competing-risks framework. Patient characteristics were comparable for all groups. Median follow-up was 65.2 months [95 % confidence interval (CI) 50.9–79.5] in the HIPEC group and 34.5 months (95 % CI 21.1–47.9) in the control group. The 5-year cumulative PM incidence was 9.3 % in the HIPEC group and 42.5 % in the control group (p = 0.004). Kaplan–Meier estimated 5-year overall survival (OS) was 81.3 % in the HIPEC group versus 70.0 % in the control group (p = 0.047). No operative death occurred. Grade 3–4 [National Cancer Institute Common Terminology Criteria for Adverse Events (NCI–CTCAE) version 4] morbidity rates were 18.2 % in the HIPEC group and 25 % in controls (p = 0.75). At multivariate analysis, HIPEC correlated to lower PM cumulative incidence [hazard ratio (HR) 0.04, 95 % CI 0.01–0.31; p = 0.002], and better OS (HR 0.25, 95 % CI 0.07–0.89; p = 0.039) and progression-free survival (HR 0.31, 95 % CI 0.11–0.85; p = 0.028). Adjuvant HIPEC may benefit CRC patients at high-risk for peritoneal failure. These results warrant confirmation in phase III trials.
- Published
- 2016
5. Noninvasive Molecular Monitoring in Multiple Myeloma Patients Using Cell-Free Tumor DNA: A Pilot Study
- Author
-
Giulia, Biancon, Silvia, Gimondi, Antonio, Vendramin, Cristiana, Carniti, and Paolo, Corradini
- Subjects
Adult ,Gene Rearrangement ,Male ,Plasma Cells ,Reproducibility of Results ,Pilot Projects ,Sequence Analysis, DNA ,Middle Aged ,Prognosis ,Circulating Tumor DNA ,Molecular Diagnostic Techniques ,Humans ,Female ,Immunoglobulin Heavy Chains ,Multiple Myeloma ,Aged - Abstract
Novel treatments for multiple myeloma (MM) have increased rates of complete response, raising interest in more accurate methods to evaluate residual disease. Cell-free tumor DNA (cfDNA) analysis may represent a minimally invasive approach complementary to multiparameter flow cytometry (MFC) and molecular methods on bone marrow aspirates. A sequencing approach using the Ion Torrent Personal Genome Machine was applied to identify clonal IGH gene rearrangements in tumor plasma cells (PCs) and in serial plasma samples of 25 patients with MM receiving second-line therapy. The same clonal IGH rearrangement identified in tumor PCs was detected in paired plasma samples, and levels of IGH cfDNA correlated with outcome and mirrored tumor dynamics evaluated using conventional laboratory parameters. In addition, IGH cfDNA levels reflected the number of PCs enumerated by MFC immunophenotyping even in the complete response context. Patients determined by MFC to be free of minimal residual disease were characterized by low frequencies of tumor clonotypes in cfDNA and longer survival. This pilot study supports the clinical applicability of the noninvasive monitoring of tumor levels in plasma samples of patients with MM by IGH sequencing.
- Published
- 2018
6. Analysis of the genomic and transcriptomic landscape of chemoresistant multiple myeloma
- Author
-
Cristiana Carniti, Michele Cavo, Vittorio Montefusco, Marina Martello, Chiara De Philippis, Giulia Biancon, Tina Bagratuni, Silvia Gimondi, Carolina Terragna, Filippo Bagnoli, Meletios A. Dimopoulos, Andrea Devecchi, Antonio Vendramin, Loris De Cecco, Paolo Corradini, Bachisio Ziccheddu, Efstathios Kastritis, Maria Luisa Sensi, Niccolo Bolli, Matteo Dugo, and Francesco Maura
- Subjects
Transcriptome ,Cancer Research ,Oncology ,business.industry ,Medicine ,Hematology ,Computational biology ,business ,medicine.disease ,Multiple myeloma - Published
- 2019
7. Analysis of the genomic landscape of multiple myeloma highlights novel prognostic markers and disease subgroups
- Author
-
Nikhil C. Munshi, Sarah O’Meara, Cristiana Carniti, Paolo Corradini, Hervé Avet-Loiseau, Giulia Biancon, Elli Papaemmanuil, Efstathios Kastritis, Silvia Gimondi, Niccolo Bolli, V. Sathiaseelan, Peter J. Campbell, Keiran Raine, Meletios A. Dimopoulos, Moritz Gerstung, Matahi Moarii, Tina Bagratuni, Chiara De Philippis, Philippe Moreau, Adam Butler, Jon W. Teague, Kenneth C. Anderson, Laura Mudie, Francesco Maura, Yu-Tzu Tai, Yang Li, Stephane Minvielle, Department of Oncology and Onco-Hematology [Milan, Italy], University of Milan, Department of Medical Oncology and Hematology [Milan, Italy] ( Fondazione IRCCS ), Fondazione IRCCS Istituto Nazionale dei Tumori, Cancer Genome Project, Wellcome Trust Sanger Institute, Epidemiology and Biostatistics [New York, NY, USA], Memorial Sloane Kettering Cancer Center [New York], Jerome Lipper Center for Multiple Myeloma Research [Boston, MA, USA] ( LeBow Institute for Myeloma Therapeutics ), Dana-Farber Cancer Institute [Boston]-Harvard Medical School [Boston] ( HMS ), Computational and Cancer Biology [Cambridge, UK], European Bioinformatics Institute [Cambridge, UK], Department of Clinical Therapeutics [Athens, Greece], National and Kapodistrian University of Athens, Département d'Hématologie [CHU Nantes], Centre hospitalier universitaire de Nantes ( CHU Nantes ), Integrative oncogenomics of multiple myeloma pathogenesis and progression ( CRCINA - Département NOHMAD - Equipe 11 ), Centre de recherche de Cancérologie et d'Immunologie / Nantes - Angers ( CRCINA ), Université d'Angers ( UA ) -Université de Nantes ( UN ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ) -Institut de Recherche en Santé de l'Université de Nantes ( IRS-UN ) -Centre hospitalier universitaire de Nantes ( CHU Nantes ) -Université d'Angers ( UA ) -Université de Nantes ( UN ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ) -Institut de Recherche en Santé de l'Université de Nantes ( IRS-UN ) -Centre hospitalier universitaire de Nantes ( CHU Nantes ), Institut Universitaire du Cancer de Toulouse - Oncopole ( IUCT Oncopole - UMR 1037 ), Université Paul Sabatier - Toulouse 3 ( UPS ) -CHU Toulouse [Toulouse]-Institut National de la Santé et de la Recherche Médicale ( INSERM ), This work was supported by a grant from NIH PO1-155258 (NCM, PJC, KCA, HAL and SM), RO1-124929 (NCM) P50-100007 (KCA, NCM) and by the Wellcome Trust (grant reference 077012/Z/05/Z). NB was funded by a MFAG grant n. 17658 from the Associazione Italiana Ricerca sul Cancro (AIRC). FM was supported by the Associazione Italiana Leucemie e Linfomi (AIL) and by the Società Italiana di Ematologia Sperimentale (SIES). PJC is personally funded through a Wellcome Trust senior clinical research fellowship., Department of Medical Oncology and Hematology [Milan, Italy] (Fondazione IRCCS), The Wellcome Trust Sanger Institute [Cambridge], Jerome Lipper Center for Multiple Myeloma Research [Boston, MA, USA] (LeBow Institute for Myeloma Therapeutics), Harvard Medical School [Boston] (HMS)-Dana-Farber Cancer Institute [Boston], National and Kapodistrian University of Athens (NKUA), Centre hospitalier universitaire de Nantes (CHU Nantes), Integrative Oncogenomics of Multiple Myeloma Pathogenesis and Progression (CRCINA-ÉQUIPE 11), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), Institut Universitaire du Cancer de Toulouse - Oncopole (IUCT Oncopole - UMR 1037), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-CHU Toulouse [Toulouse]-Institut National de la Santé et de la Recherche Médicale (INSERM), Bernardo, Elizabeth, Università degli Studi di Milano = University of Milan (UNIMI), Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Centre Hospitalier Universitaire de Toulouse (CHU Toulouse)-Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Toulouse [Toulouse]-Université Toulouse III - Paul Sabatier (UT3), and Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)
- Subjects
0301 basic medicine ,Male ,Cancer Research ,DNA Copy Number Variations ,Genotype ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Disease ,Computational biology ,Gene mutation ,Biology ,Translocation, Genetic ,Article ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,PRDM1 ,medicine ,Biomarkers, Tumor ,Humans ,Gene ,Multiple myeloma ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,High-Throughput Nucleotide Sequencing ,Hematology ,Genomics ,Middle Aged ,medicine.disease ,Prognosis ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Proteasome inhibitor ,Female ,Positive Regulatory Domain I-Binding Factor 1 ,Multiple Myeloma ,medicine.drug - Abstract
International audience; In multiple myeloma, next generation sequencing (NGS) has expanded our knowledge of genomic lesions, and highlighted a dynamic and heterogeneous composition of the tumor. Here, we used NGS to characterize the genomic landscape of 418 multiple myeloma cases at diagnosis and correlate this with prognosis and classification. Translocations and copy number changes (CNAs) had a preponderant contribution over gene mutations in defining the genotype and prognosis of each case. Known and novel independent prognostic markers were identified in our cohort of proteasome inhibitor and IMiD-treated patients with long follow-up, including events with context-specific prognostic value, such as deletions of the PRDM1 gene. Taking advantage of the comprehensive genomic annotation of each case, we used innovative statistical approaches to identify potential novel myeloma subgroups. We observed clusters of patients stratified based on the overall number of mutations and number/type of CNAs, with distinct effects on survival, suggesting that extended genotype of multiple myeloma at diagnosis may lead to improved disease classification and prognostication.
- Published
- 2017
8. Pharmacologic Inhibition of JAK1/JAK2 Signaling Reduces Experimental Murine Acute GVHD While Preserving GVT Effects
- Author
-
Cristiana Carniti, Antonio Vendramin, Silvia Gimondi, Jacopo Mariotti, Camilla Recordati, Paolo Corradini, Anisa Bermema, and Davide Confalonieri
- Subjects
Cancer Research ,Ruxolitinib ,T-Lymphocytes ,medicine.medical_treatment ,Graft vs Host Disease ,Bone Marrow Cells ,Hematopoietic stem cell transplantation ,Disease ,Major histocompatibility complex ,Mice ,Chemokine receptor ,In vivo ,Nitriles ,medicine ,Animals ,Humans ,Transplantation, Homologous ,biology ,Kinase ,business.industry ,Hematopoietic Stem Cell Transplantation ,Cell Differentiation ,Janus Kinase 1 ,Janus Kinase 2 ,Transplantation ,Disease Models, Animal ,Pyrimidines ,surgical procedures, operative ,Oncology ,Immunology ,biology.protein ,Pyrazoles ,business ,Signal Transduction ,medicine.drug - Abstract
Purpose: Immune-mediated graft-versus-tumor (GVT) effects can occur after allogeneic hematopoietic stem cell transplantation (HSCT), but GVT is tightly linked to its main complication, graft-versus-host disease (GVHD). Strategies aimed at modulating GVHD, while maintaining the GVT effect, are needed to improve the cure rate of transplant. Given the emerging role of Janus-activated kinase (JAK) signaling in lymphoproliferative and myeloproliferative diseases and its established function at dictating T-cell differentiation, we postulated that JAKs might be potential therapeutic targets through a pharmacologic approach. Experimental Design: We examined the effect of JAK1/JAK2 modulation by ruxolitinib in a mouse model of fully MHC mismatched bone marrow transplant comprising in vivo tumor inoculation. Results: JAK1/JAK2 inhibition by ruxolitinib improved both overall survival (P = 0.03) and acute GVHD pathologic score at target organs (P ≤ 0.001) of treated mice. In addition, treatment with ruxolitinib was associated with a preserved GVT effect, as evidenced by reduction of tumor burden (P = 0.001) and increase of survival time (P = 0.01). JAK1/JAK2 inhibition did not impair the in vivo acquisition of donor T-cell alloreactivity; this observation may account, at least in part, to the preserved GVT effect. Rather, JAK1/JAK2 inhibition of GVHD was associated with the modulation of chemokine receptor expression, which may have been one factor in the reduced infiltration of donor T cells in GVHD target organs. Conclusions: These data provide further evidence that JAK inhibition represents a new and potentially clinically relevant approach to GVHD prevention. Clin Cancer Res; 21(16); 3740–9. ©2015 AACR.
- Published
- 2015
9. Graft Monocytic Myeloid-Derived Suppressor Cell Content Predicts the Risk of Acute Graft-versus-Host Disease after Allogeneic Transplantation of Granulocyte Colony-Stimulating Factor–Mobilized Peripheral Blood Stem Cells
- Author
-
Cristiana Carniti, Paolo Corradini, Antonio Vendramin, Silvia Gimondi, Anisa Bermema, Paolo Longoni, and Sara Rizzitano
- Subjects
Adult ,Male ,Allogeneic transplantation ,Adolescent ,medicine.medical_treatment ,Graft vs Host Disease ,Transplants ,Hematopoietic stem cell transplantation ,Graft-versus-host disease ,Monocytes ,Predictive Value of Tests ,Risk Factors ,Granulocyte Colony-Stimulating Factor ,medicine ,Humans ,Prospective Studies ,Aged ,Peripheral Blood Stem Cell Transplantation ,Transplantation ,business.industry ,Hematology ,Middle Aged ,Allografts ,medicine.disease ,Hematopoietic Stem Cell Mobilization ,Granulocyte colony-stimulating factor ,Granulocyte colony-stimulating factor mobilization ,Haematopoiesis ,surgical procedures, operative ,Myeloid-derived suppressor cells ,Allogeneic hematopoietic stem cell transplantation ,Acute Disease ,Immunology ,Myeloid-derived Suppressor Cell ,Female ,Stem cell ,Unrelated Donors ,business - Abstract
Myeloid-derived suppressor cells (MDSCs) are powerful immunomodulatory cells that in mice play a role in infectious and inflammatory disorders, including acute graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation. Their relevance in clinical acute GVHD is poorly known. We analyzed whether granulocyte colony-stimulating factor (G-CSF) administration, used to mobilize hematopoietic stem cells, affected the frequency of MDSCs in the peripheral blood stem cell grafts of 60 unrelated donors. In addition, we evaluated whether the MDSC content in the peripheral blood stem cell grafts affected the occurrence of acute GVHD in patients undergoing unrelated donor allogeneic stem cell transplantation. Systemic treatment with G-CSF induces an expansion of myeloid cells displaying the phenotype of monocytic MDSCs (Linlow/negHLA-DR−CD11b+CD33+CD14+) with the ability to suppress alloreactive T cells in vitro, therefore meeting the definition of MDSCs. Monocytic MDSC dose was the only graft parameter to predict acute GVHD. The cumulative incidence of acute GVHD at 180 days after transplantation for recipients receiving monocytic MDSC doses below and above the median was 63% and 22%, respectively (P = .02). The number of monocytic MDSCs infused did not impact the relapse rate or the transplant-related mortality rate (P > .05). Although further prospective studies involving larger sample size are needed to validate the exact monocytic MDSC graft dose that protects from acute GVHD, our results strongly suggest the modulation of G-CSF might be used to affect monocytic MDSCs graft cell doses for prevention of acute GVHD.
- Published
- 2014
- Full Text
- View/download PDF
10. Disease Monitoring in Multiple Myeloma Patients Using Liquid Biopsy and Next Generation Sequencing of IGH Gene Rearrangements
- Author
-
Cristiana Carniti, Giulia Biancon, Antonio Vendramin, Vittorio Montefusco, Sofia Cannara Malan, Paolo Corradini, Sara Rizzitano, Silvia Gimondi, Silvia Zaninelli, and Anisa Bermema
- Subjects
education.field_of_study ,Pathology ,medicine.medical_specialty ,Immunology ,Population ,Cell Biology ,Hematology ,Gene rearrangement ,Ion semiconductor sequencing ,Biology ,Amplicon ,medicine.disease ,Biochemistry ,Minimal residual disease ,Molecular biology ,law.invention ,law ,medicine ,Liquid biopsy ,education ,Polymerase chain reaction ,Multiple myeloma - Abstract
Background: Current criteria used to assess response lag behind the extraordinary evolution in the treatment of multiple myeloma (MM) patients (pts), and more sensitive techniques are being explored to detect true minimal residual disease (MRD) for new complete remission (CR) definitions. In recent years, next generation sequencing (NGS) technologies have emerged. NGS of immunoglobulin (IgH) gene rearrangements are very sensitive and also allow the identification of small subclonal population that can be monitored over time during treatment, something not possible with flow cytometry or PCR. However, the patchy pattern of bone marrow infiltration observed in MM leads to some degree of uncertainty regarding MRD-negative results, irrespectively of the technique adopted. This highlights the value of applying "liquid biopsy" as a non-invasive strategy for monitoring MRD through the analysis of circulating cell-free tumor DNA (ctDNA). The objective of the current study was to measure residual tumor burden in sequential plasma samples of a cohort of MM pts by NGS of the IgH gene rearrangements. Methods: We retrospectively analyzed 14 MM pts homogeneously treated between 2011 and 2015 with all clinical data available. We obtained serial tumor and plasma samples at diagnosis and at specified time points during treatment cycles and up to 24 months of follow-up. Genomic DNA (gDNA) was extracted from immunomagnetically selected CD138+ plasma cells at diagnosis (n=14). ctDNA was extracted from 500uL of plasma (Qiagen) at diagnosis (n=14) and at follow-up time points (n=58). IgH gene rearrangements were amplified, quality assessed (Agilent hsDNA kit) and sequenced on Ion Torrent PGM as previously described (Gimondi et al., ASH 2015). Raw reads were filtered for quality, length (>250bp) and presence of both forward and reverse primers. Reads were subsequently aligned using IgBlast against IMGT germline database and aggregated into clonotypes based on identity of CDR3, V and J segments (MigMap). Post-processing analyses were performed using VDJtools and customized R scripts. Results: PCR products quality assessment from ctDNA amplification of the entire IgH-VDJ region revealed the presence of both short (150-250bp) and long amplicons (310-360bp). Raw reads were subjected to filtering using our custom bioinformatic workflow to retain only complete IgH-VDJ gene rearrangements and discard low-quality reads. Three pts could not be evaluated due to low quality sequencing reads in all samples. At least 3 follow-up time points were available for all the remaining 11 pts whereas 6 pts had 4 time points. At diagnosis, both plasma and tumor samples revealed a high level of heterogeneity (range 1980-7753 clonotypes) with only a small fraction of shared clonotypes (346±262, mean±SD). Among the shared ones, the clonotype with the highest frequency in plasma corresponded to the tumor-associated one identified in CD138+ cells. Interestingly, in the plasma of 3 pts, additional clonotypes were detected at relatively high frequencies (range 1-16%) suggesting the presence of subclones. IgH-NGS at follow-up time points revealed that the clonotype identified at diagnosis (range 4-31% of total reads) could be easily tracked over time in plasma samples, at frequencies as low as 0.00001%. Frequencies of the tumor-associated IgH gene rearrangement in plasma showed a patient-specific modulation and reflected the tumor burden assessed according to the International Myeloma Working Group-Uniform Response Criteria. At the time of CR, the tumor-associated clonotype was undetectable in the plasma of pts who would not subsequently relapse. In patients that would lately experience progressive disease, the tumor specific clonotype was still detectable at low frequencies (range 0.00001-0.03%) in all plasma samples suggesting that liquid biopsy can be used for MRD monitoring. Conclusions: Despite the limited number of pts and follow-up samples analyzed, we demonstrate that NGS of IgH gene rearrangements from ctDNA can be used for MM disease monitoring, thus representing a non-invasive alternative strategy for clinical management. The analysis of retrospectively collected plasma samples revealed that ctDNA quality is essential for a NGS characterization of IgH gene rearrangements. Plasma samples collection and processing represent critical steps that need to be considered designing prospective liquid biopsy studies. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
11. Synergistic Anti-Tumor Efficacy of BET Inhibitors JQ1 and Otx-015 in Combination with Dasatinib in Preclinical Models of T-Cell Lymphomas
- Author
-
Silvia Gimondi, Antonio Vendramin, Cristiana Carniti, Giulia Biancon, Alessandra Cavanè, Sara Rizzitano, Marco Piazzoni, Anna Dodero, Sofia Cannara Malan, and Paolo Corradini
- Subjects
Cell cycle checkpoint ,medicine.diagnostic_test ,T cell ,Immunology ,030206 dentistry ,Cell Biology ,Hematology ,Biology ,Cell cycle ,Pharmacology ,Biochemistry ,Jurkat cells ,Flow cytometry ,Dasatinib ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Apoptosis ,030220 oncology & carcinogenesis ,medicine ,Tyrosine kinase ,medicine.drug - Abstract
Background: Approximately 50% of patients with peripheral T-cell lymphoma (PTCL) enter long-term remission after standard chemotherapy and stem cell transplantation. Patients who do not respond to chemotherapy have few treatment options highlighting the critical need for new effective and targeted therapeutics. Aberrant T cell receptor (TCR) and tyrosine kinase (TK) signaling have been described in PTCL (Agostinelli 2014;Netchiporouka 2014). Single-agent TK inhibitors (TKIs) have significantly improved patient outcomes across multiple tumor subtypes. However, TKI therapy is rarely curative. The recent discovery of a subgroup of PTCL characterized by high levels of GATA3 and c-Myc expression and poor prognosis (Iqbal 2014; Manso 2016), establishes the rationale of targeting c-Myc in PTCLs. Based on the demonstration that pharmacologic inhibition of c-Myc is achievable through targeting bromodomain and extra terminal (BET) family of chromatin adapters, the therapeutic potential of BET inhibition was assessed in a panel of T cell lymphoma and leukemia cell lines. Since expression of c-Myc is regulated by the TCR, we also hypothesized that simultaneous targeting of c-Myc and TCR would significantly enhance the antiproliferative effects of BET inhibitors (BETis) and TKI alone in preclinical models of PTCL. Methods: Five T-cell lymphoma and leukemia cell lines (Jurkat, HD-MAR-2, Karpas 299, Sup-T1, HH) were incubated with escalating doses of JQ1 (a small-molecule BETi with the highest affinity for BRD4) and OTX-015 (a BETi with a broader affinity for BRD2, BRD3, BRD4) and the tyrosine-kinase-inhibitor Dasatinib. Analysis of cell viability, cell cycle distribution, apoptosis and mitochondrial depolarization was performed using flow cytometry. Effects of treatments were assessed using gene expression profiling (GEP) and western blotting (WB). Combinations were evaluated using the Chou-Talalay Combination Index (CI), calculated with CompuSyn software (CompuSyn Inc, Paramus, NJ). Results: JQ1 and OTX-015 show antiproliferative activity with IC50 at nanomolar concentrations in all cell lines. As assessed determining viable cells by PI exclusion and flow cytometry, JQ1 and OTX-015 are similarly active in a dose-dependent manner in all cell lines. To understand the activity of JQ1 and OTX-015, we analyzed cell-cycle distribution using flow cytometry. JQ1 and OTX-015 induce a cell cycle arrest with G1-phase accumulation and decrease S-phase with the exception of SUPT1 cells that are characterized by a cell cycle arrest in G2-phase. Minimal increase in the sub-G1 population is observed in all cell lines, suggesting that JQ1 and OTX-015 mainly exert a cytostatic effect. We then examined GATA3 and c-Myc protein levels in all cell lines: varying amounts of GATA3 and c-Myc proteins were observed but a strong correlation between GATA3 and c-Myc expression was detected. After JQ1 and OTX-015 exposure, c-Myc protein level decrease in all cell lines apart from SUP-T1 cell line. Here c-Myc level do not change significantly upon BETis exposure, suggesting that BETis target other pathways relevant for SUP-T1 survival. Dasatinib efficiently inhibits the proliferation in all cell lines at micromolar concentrations in a dose-dependent manner. Dasatinib induces G0/G1-phase arrest and an increase in sub-G1 population indicating a modest induction of apoptosis confirmed by caspase-9 activation and mitochondrial depolarization. Compared to all single agents, combined treatments with sub-optimal concentrations of Dasatinib and JQ1 or OTX-015 exert synergistic lethal activity against all tested cell lines (C.I. Conclusions: The experiments presented here support the combination of BET inhibitors with the TK inhibitor Dasatinib for PTCLs. Our data suggest a synergistic interaction for the combination of both BETis and Dasatinib in vitro. Mechanistically, combined treatments exert synergistic anti-tumor effects in all cell lines through growth inhibitory effects, direct induction of cell death by promotion of caspase-dependent apoptosis and mitochondrial depolarization. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
12. Identification of Clonal Igh Gene Rearrangements By High-Throughput Sequencing of Cell Free DNA in Multiple Myeloma Patients
- Author
-
Paolo Corradini, Silvia Gimondi, Alessandra Cavanè, Cristiana Carniti, Vittorio Montefusco, Biancon Giulia, Antonio Vendramin, and Anisa Bermema
- Subjects
Sanger sequencing ,Immunoglobulin gene ,Immunology ,Cell Biology ,Hematology ,Gene rearrangement ,Ion semiconductor sequencing ,Biology ,Amplicon ,Biochemistry ,Molecular biology ,Minimal residual disease ,DNA sequencing ,law.invention ,symbols.namesake ,law ,symbols ,Polymerase chain reaction - Abstract
Background: The achievement of complete remission is an important therapeutic goal and prognostic factor for overall survival in patients with B-cell malignancies. Molecular monitoring of disease with PCR-based strategies is used to assess the depth of treatment response, detect minimal residual disease (MRD), and identify patients at increased risk of relapse. Immunoglobulin heavy chain (IGH) gene rearrangements are used as molecular marker in approximately 80% of lymphoma and multiple myeloma (MM) patients since they represent lineage-specific markers and the third complementarity determining region (CDR3) is unique to each clone. We and others have demonstrated that next generation sequencing (NGS) technologies provide the opportunity to identify and quantify clonotypes with consensus primers combining the benefits of high sensitivity and universal applicability thus overcoming ASO-PCR and RQ-PCR limitations. Very recently, NGS has been applied to characterize the tumor-specific VDJ recombination of the immunoglobulin genes in the serum of patients with diffuse large B-cell lymphoma and used as a novel non-invasive strategy to predict clinical disease recurrence after first-line treatment (Roschewski et al., Lancet Oncology 2015). The present study was designed to assess whether the Ion Torrent Personal Genome Machine (IT-PGM)-based sequencing and analysis we established (Gimondi et al., ASH 2014), could be used to detect circulating tumor DNA encoding the clonal immunoglobulin gene sequence in the plasma of patients with Multiple Myeloma. Methods: Genomic DNA (gDNA) was extracted from CD138+ plasma cells immunomagnetically selected from the bone marrow blood of six MM patients and amplified using seven different family-specific IgH-V primers and a consensus JH primer (Voena et al., Leukemia 1997). IgH clonality was assessed by Sanger sequencing in order to define the patient specific DNA rearrangement. Plasma samples of the six MM patients were collected and cell free DNA (cfDNA) extracted (Qiagen). The total amount of cfDNA was estimated by fluorometric measurement (median 105 ng, range 37-171 ng).For library construction and NGS, paired samples of gDNA (500ng) and cfDNA (at least 37ng) were amplified as previously described (Gimondi et al., ASH 2014). PCR products were evaluated for quality and length by high-sensitivity dsDNA chips (Agilent).PCR amplicons were barcoded, pooled and sequenced on the Ion Torrent PGM.NGS data were analyzed by using the IMGT-High V-Quest web server tool and the statistical software R. Results: Rearranged IGHV-D-J loci from cfDNA of each MM patient could be amplified using 7 different IGHV family primers and a consensus JH primer, despite the limited abundance of DNA recovered from plasma samples. PCR products were sequenced on an IT-PGM 316 chip, yielding at least 110K reads per sample (mean 280K reads) with an average coverage of 130x (at least 50x). Clonal IgH sequences were quantified as a fraction of the complete IGHV-D-J rearranged reads. The clonality of our samples was assessed by determining the percentage of reads identical to the most abundant CDR3 sequence in each sample. The number of clonal sequences corresponding to the highest expressed IGH clonotype in gDNA was consistent with those identified in cfDNA samples (range 74%-82% and range 65%-73% respectively). Furthermore, the clonotypes identified by high-throughput sequencing of gDNA and cfDNA samples demonstrated a 100% sequence identity with the patient-specific IGHV-D-J clonal rearrangement identified by Sanger sequencing. Conclusions: We demonstrate that next generation sequencing of cfDNA from the plasma of Multiple Myeloma patients is feasible, accurate, and sensitive in identifying the tumor-derived VDJ recombination of the immunoglobulin genes without prior knowledge of the tumor clonotype and might represent an alternative when bone marrow biopsies are unavailable. Moreover, given the patchy bone infiltration of malignant plasma cells, cfDNA analysis is a non-invasive approach that might give a more precise quantification of disease burden. In addition, NGS analysis of the IGHV-D-J rearranged sequences provides a deep and detailed characterization of the patient immune repertoire, thus possibly allowing clonal evolution evaluations and monitoring of MRD in follow-up samples. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
13. Genomic Landscape and Its Prognostic Implications in Multiple Myeloma Using a Targeted Sequencing Approach
- Author
-
Silvia Gimondi, Yang Li, Hervé Avet-Loiseau, Stephane Minvielle, Adam S. Sperling, Peter J. Campbell, Kenneth C. Anderson, V. Sathiaseelan, Niccolo Bolli, and Nikhil C. Munshi
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Genetics ,Point mutation ,Immunology ,Single-nucleotide polymorphism ,Genomics ,Cell Biology ,Hematology ,Gene mutation ,Biology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,medicine ,KRAS ,Multiple myeloma ,Exome sequencing - Abstract
Next-generation sequencing (NGS) studies have shown that multiple myeloma is a heterogeneous disease with a complex subclonal architecture and few recurrently mutated genes. Only a minority of variants are potentially actionable and studies on their prognostic value are lacking. Therefore, strategies to investigate the landscape of chromosomal and gene lesions of a large number of myeloma samples in a robust fashion is needed. In this study, we developed a target-enrichment strategy to streamline simultaneous analysis of gene mutations, copy number changes and IGH translocations in multiple myeloma in a high-throughput fashion using NGS. We designed Agilent SureSelect cRNA pull down baits to target 246 genes implicated in myeloma, in lymphoid malignancies or other cancers, 2538 single nucleotide polymorphisms to detect copy number and allelic ratio at the single-gene and whole-genome level, and we tiled the whole IGH locus to detect IGH translocations and V(D)J rearrangements. As a pilot, we sequenced 13 myeloma cell lines and 10 control haematopoietic cells lines and validated its sensitivity and specificity. We have next applied this baitset to unmatched DNA from CD138-purified plasma cells from 426 patients at diagnosis, including 51 matched samples from a previous whole exome sequencing dataset. We sequenced at an average depth of 329x with 77% of the target region covered at >30x using Hiseq2000 machines (Illumina Inc.). We applied algorithms developed in-house to detect point mutations, insertion-deletions and IGH translocations, filtering out potential artifacts and germline variants. We then annotated as "oncogenic" all variants previously reported as somatic in cancer by the COSMIC database. 418/426 patients had at least one variant, and overall we identified 2207 variants of which 667 were oncogenic. 212/246 evaluated genes were mutated at least once, but only 102 had at least one oncogenic variant. Furthermore, 417/667 (63%) oncogenic variants were accounted for by the top 8 driver genes previously identified (KRAS, NRAS, TP53, FAM46C, BRAF, DIS3, TRAF3, SP140). Additional findings of interest included clustered mutations in SF3B1, EGR1, IRF4, but in We then looked at prognostic models. While TP53 mutations had the strongest effect both on overall survival (OS) and progression free survival (PFS), we found a favorable impact of TRAF3 mutations on PFS, and a negative one of NRAS and SP140. More importantly, pairwise interactions served as a starting ground for a rationale subgroup analysis. For example, while FAM46C mutations had no effect on OS across the whole cohort, they predicted better survival in patients without IGH translocations (median 94 months for WT FAM46C vs. not reached for MUT, p=0.042 log-rank test). Similarly, we found a different magnitude of the negative impact of TP53 mutations on OS based on karyotype. Survival was shortened more than 8-fold in cases with both an IGH translocation and a TP53 mutation (median 89 months for WT TP53 vs. 11 for MUT, p=2e-10), while this effect was much less in the absence of an IGH translocation (94 months for WT vs. 50 for MUT, p=0.02). In conclusion, the large sample size and extent of sequencing provides further insight into the genomic landscape of myeloma and how this impacts the clinical phenotype confirming the utility of the targeted sequencing to both understand the biology as well as its clinical application. Disclosures Campbell: 14M genomics: Other: Co-founder and consultant.
- Published
- 2015
14. Pharmacologic Inhibition of JAK1/JAK2 Signaling Protects from Acute GvHD While Preserving Gvt in Mice
- Author
-
Silvia Gimondi, Anisa Bermema, Cristiana Carniti, Davide Confalonieri, Paolo Corradini, Antonio Vendramin, Jacopo Mariotti, and Camilla Recordati
- Subjects
Myeloid ,business.industry ,medicine.medical_treatment ,T cell ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Biochemistry ,Transplantation ,medicine.anatomical_structure ,Immune system ,Cytokine ,medicine ,Bone marrow ,business ,CD8 - Abstract
Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents a potential curative strategy for patients with hematological malignancies. Despite recent improvements in transplantation procedurs and supportive care, acute graft versus host disease (aGvHD) remains the most significant barrier to the success of allo-HSCT. In this contest, strategies directed against GvHD adversely affect survival because they increase malignancy relapse and infections. Approaches aimed at separating graft-versus-tumor (GvT) effect from GvHD are warranted, but difficult to achieve because of their shared biology. Given that JAK signaling dictates T cell differentiation, we postulated that JAKs might be potential therapeutic targets in allo-HSCT through a pharmacological approach. The relative importance of JAK2 signaling in myeloid and lymphoid malignancies drew our attention to the usage of INCB18424 (Ruxolitinb), a JAK1/JAK2 specific inhibitor that was recently shown to be an effective treatment for patients with myelofibrosis. We tested our hypothesis in a mouse model of aGvHD in order to abrogate GvHD, while maintaining a GvT effect. Methods: A major histocompatibility complex (MHC) mismatched HSCT mouse model was set up. Lethally irradiated BALB/c mice received spleen (SC) and bone marrow (BM) cells from donors C57BL/6 (B6) mice, and were treated with INCB18424 for 14 days at the dose of 90mg/kg/day (INC90), 45mg/kg/day (INC45) or 22.5mg/kg/day (INC22.5). Syngeneic transplants (B6-B6) and BALB/c recipients treated with B6 BM cells only were also used. To determine the GvT activity, allo-HSCT recipients were co-injected with either a B-cell lymphoma cell line (A20) or a myeloid leukemia cell line (RMB-1) and treated or not with INCB18424 at the dose of 45mg/kg/day. Mice were monitored for overall survival (OS) and weight loss. GvHD was histologically scored in tissues harvested on day 14, 30 and 60 and cytokine production by ELISA. Immune reconstitution and tumor cells were monitored by flow cytometry. Results: Significantly less GvHD, as determined by survival (INC45, p Conclusions: The inhibition of Jak/STAT signalingusing the sensitive and specific inhibitor of Jak1/Jak2, INCB18424 conferred effective protection from lethal acute GvHD in a MHC mismatched HSCT mouse model while sparing the GvT effect. Our work provides novel evidences of the mechanisms implied in INCB18424-mediated prevention of acute GvHD, demonstrating a significant inhibition of T cell trafficking into GvHD target tissues associated with reduced expression of CXCR3. We expect that these experimental observations may be readily translated into a prospective clinical trial evaluating INCB18424 (Ruxolitinb) for aGvHD prophilaxis after allo-HSCT for patients with hematologic malignancies. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
15. a Novel Method for the Detection of Minimal Residual Disease in B-Cell Malignancies: Ion Semiconductor Sequencing for the Evaluation of Igh Gene Rearrangements
- Author
-
Antonio Vendramin, Paolo Corradini, Cristiana Carniti, Giulia Biancon, Silvia Gimondi, and Alessandra Cavanè
- Subjects
Sanger sequencing ,Immunology ,Cell Biology ,Hematology ,Computational biology ,Ion semiconductor sequencing ,Biology ,Biochemistry ,Minimal residual disease ,DNA sequencing ,law.invention ,symbols.namesake ,law ,Allele-specific oligonucleotide ,Multiplex polymerase chain reaction ,symbols ,Primer (molecular biology) ,Polymerase chain reaction - Abstract
Background: Minimal residual disease (MRD) detection is of high clinical relevance in patients with B-cell malignancies and is generally a surrogate parameter to evaluate treatment response and long-term prognosis. IgH gene rearrangements can be used as molecular marker in approximately 80% of lymphoma and myeloma patients since they represent lineage-specific markers and the complementarity determining region 3 (CDR3) is unique to each clone. To date, allele specific oligonucleotide polymerase chain reaction (ASO-PCR) and real-time quantitative polymerase chain reaction (RQ-PCR) are considered the most sensitive and widely applicable methods for MRD detection. A major disadvantage of ASO-PCR and RQ-PCR assays, is the use of specific primers and probes for every individual patient. Clone-specific primers and probes are not only expensive but also time-consuming to design and to test, which limits their wide applicability in the clinical setting. The recent major improvements in next generation sequencing (NGS) technologies, provide the opportunity to identify and quantify clonotypes with consensus primers combining the benefits of high sensitivity and universal applicability. The present work was designed to overcome ASO-PCR and RQ-PCR limitations by developing a feasible method for rearranged IgH genes amplification, NGS and analysis using Ion Torrent Personal Genome Machine (IT-PGM). Methods: To define a multiplex PCR protocol, DNA from 7 CLL patients, previously shown to bare a family specific clonal VDJ rearrangement, was amplified with a pool of the seven different family-specific IgH-V primers, and a consensus JH primer (Voena et al., Leukemia 1997). After Sanger sequencing, results were compared to the ones obtained with singleplex PCR protocol. Once validated, the multiplex PCR protocol was used to amplify DNA from patients and serially diluted (up to 10-8 ) DNA from Namalwa cell line (bearing a known IgH rearrangement) and subsequently sequenced on the IT-PGM using the 316 Ion-chip. NGS data were analyzed by using the IMGT-High V-Quest web server tool and the statistical software R. RQ-PCR was used to quantify the specific VDJ rearrangement in the serially diluted Namalwa DNA solutions and in DNA from patients as previously described (Farina et al, Haematologica 2009). RQ-PCR data were analyzed through a relative quantification procedure. Results: The multiplex PCR reactions we have tested, demonstrated the same specificity as the standard singleplex PCR protocol and therefore was used to construct the DNA library required for IT-PGM-based sequencing. The IT-PGM sequencing output is represented by at least 400000 reads per sample with a minimum average coverage of the VDJ repertoire of 500x. The IMGT-High V-quest tool allows a user-friendly web based analysis and a deep molecular characterization of the IgH recombinatorial repertoire. Namalwa clonal CRD3 sequences were detected up to a dilution of 10-5 without the need for specific CDR3 primers. Comparability of NGS and ASO RQ-PCR results was assessed. The use of CDR3 specific primers, along with the specific IgH-V family fluorescent probe, enabled the identification of clonal VDJ rearrangements with a sensitivity up to 10-5 (2/3 replicates) and 10-6 (just 1/3 replicates) in Namalwa Cell Line. Similar results were obtained when we characterized the IgH recombination repertoire of two CLL patients over time. Conclusions: IgH sequencing with the IT-PGM platform showed at least the same level of sensitivity as ASO RQ-PCR, without the need for patient-specific reagents. It also allows specific and detailed molecular characterization of the clonal rearrangements and could be easily incorporated into clinical laboratories for routine testing of MRD in B-cell malignancies. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
16. Abstract 1693: The combination of romidepsin and bendamustin is synergistically cytotoxic and reverses the malignant phenotype in preclinical models of T-cell lymphoma
- Author
-
Antonio Vendramin, Cristiana Carniti, Sara Rizzitano, Silvia Gimondi, and Paolo Corradini
- Subjects
Bendamustine ,Cancer Research ,Vincristine ,Cyclophosphamide ,business.industry ,CHOP ,Pharmacology ,medicine.disease ,Lymphoma ,Romidepsin ,Oncology ,Cancer research ,Medicine ,T-cell lymphoma ,Doxorubicin ,business ,medicine.drug - Abstract
Background: Peripheral T-cell lymphomas (PTCLs) represent approximately 10-15% of all non-Hodgkin lymphomas (NHL) in the Western world, and their incidence is increasing. Cases of PTCL tend to have an aggressive clinical course, with poor patient responses to conventional chemotherapy and poor long-term survival. So far, treatment approaches have mirrored diffuse large B-cell lymphoma (DLBCL) and CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) and CHOP-like chemotherapy are commonly used despite suboptimal results. Histone deacetylase inhibitors (HDACs) have been approved for the treatment of relapsed or refractory PTCLs given their single-agent activity in these diseases. To continue to improve responses in patients with PTCL, it is important to assess the utility of romidepsin as frontline therapy and as a component of combination therapies. Aim: Interactions between romidepsin (R) and bendamustine (B), a potent cytotoxic alkylating drug active against a panel of other lymphoproliferative disorders, was investigated in in preclinical models of T-cell Lymphoma. Experimental Design: Assays for cytotoxicity on 5 different T-cell lymphoma and leukemia cell lines (Jurkat, HD-MAR2, Karpas, Sup-T1, HH), mathematical analysis for synergism (Chou-Talalay equation), flow cytometry, and the Itk-Syk transgenic mouse model (K. Pechloff, 2010) were used to explore the in vitro and in vivo activities of R and B alone and in combination in T-cell lymphoid malignancies. Results: In vitro, romidepsin and bendamustine exhibited concentration- and time-dependent cytotoxicity against all the 5 different T-cell lymphoma and leukemia cell lines. Romidepsin showed synergism when combined with bendamustine in all cell lines studied. Romidepsin also induced potent apoptosis and caspase activation when combined with bendamustine across the panel. The impact of schedule on the activity of the combination was determined by assessing cell viability after treatment with B and R as follows: (1) simultaneous exposure; (2) B pretreatment followed by exposure to R; and (3) R pretreatment followed by exposure to B. In a new mouse model of PTCL in which a status of permanent T cell activation mediated by the Itk-Syk transcript induces highly malignant PTCLs with 100% penetrance that resemble the human disease, the combination of romidepsin and bendamustine enhanced efficacy compared with either drug alone. Conclusions: Collectively, these data strongly support the potential therapeutic role of romidepsin in combination with bendamustine for PTCLs and might constitute the basis for future phase I-II clinical trials. Citation Format: Cristiana Carniti, Silvia Gimondi, Antonio Vendramin, Sara Rizzitano, Paolo Corradini. The combination of romidepsin and bendamustin is synergistically cytotoxic and reverses the malignant phenotype in preclinical models of T-cell lymphoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1693. doi:10.1158/1538-7445.AM2014-1693
- Published
- 2014
17. In Vivo Jak1/Jak2 Inhibition Protects From Acute GvHD While Maintaining Robust Anti-Tumor Activity
- Author
-
Cristiana Carniti, Silvia Gimondi, Antonio Vendramin, Davide Confalonieri, Camilla Recordati, Anisa Bermema, Jacopo Mariotti, and Paolo Corradini
- Subjects
education.field_of_study ,business.industry ,medicine.medical_treatment ,Immunology ,Population ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,surgical procedures, operative ,Cytokine ,Immune system ,medicine.anatomical_structure ,Graft-versus-host disease ,immune system diseases ,medicine ,Interleukin 12 ,Bone marrow ,education ,business ,CD8 - Abstract
Background Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative option for patients with hematological malignancies. However, its success is limited by life-threatening graft-versus-host disease (GvHD). Strategies to control GvHD are often associated with suppression of the immune system leading to the impairment of the beneficial graft versus tumor (GvT) effect. The ideal approach to prevent and treat GvHD would limit alloantigen-specific reactivity while preserving immunity against malignant cells and pathogens. This study extends our previous findings on the beneficial effect of the inhibition of JAK signaling on acute GvHD (aGvHD) (Carniti et al, ASH2012), evaluating whether INCB18424 (Ruxolitinb) treatment preserved the GvT effect in a mouse model of aGvHD. Methods A major histocompatibility complex (MHC) mismatched HSCT mouse model was set up. Lethally irradiated BALB/c mice received spleen (SC) and bone marrow (BM) cells from donors C57BL/6 (B6) mice, and were treated with INCB18424 for 14 days at the dose of 90mg/kg/day (INC90), 45mg/kg/day (INC45) or 22.5mg/kg/day (INC22.5). Syngeneic transplants (B6-B6) and BALB/c recipients treated with B6 BM cells only were also used. To determine the GvT activity, allo-HSCT recipients were co-injected with either a B-cell lymphoma cell line (A20) or a myeloid leukemia cell line (RMB-1) and treated or not with INCB18424 at the dose of 45mg/kg/day. Mice were monitored for overall survival (OS) and weight loss. GvHD was histologically scored in tissues harvested on day 14, 30 and 60 and cytokine production by ELISA. Immune reconstitution and tumor cells were monitored by flow cytometry. Results INCB18424 treatment caused significantly less GvHD when compared to untreated animals, as determined by survival (INC45, p
- Published
- 2013
18. Inhibition of Jak1/Jak2 Is More Effective Than Inhibition of Jak3 in Protecting Mice From Acute Graft-Versus-Host Disease (aGvHD) by Significantly Decreasing Alloreactive CD4+ T-Cells
- Author
-
Antonio Vendramin, Cristiana Carniti, Silvia Gimondi, Enrico Radaelli, Jacopo Mariotti, Anisa Bermema, Paolo Corradini, and Raffaella Vaccaroli
- Subjects
business.industry ,medicine.medical_treatment ,Immunology ,Immunosuppression ,Spleen ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Transplantation ,Leukemia ,Graft-versus-host disease ,medicine.anatomical_structure ,medicine ,Cytotoxic T cell ,Bone marrow ,business - Abstract
Abstract 2997 Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative treatment for patients with hematological malignancies. However, its success is limited by a life-threatening complication: the graft-versus-host disease (GvHD). Although numerous studies have described immunosuppression protocols to mitigate acute GVHD (aGvHD), novel approaches are needed. Chemokines are well known inducers of leukocyte trafficking and activation. Stimulation of the chemokine receptor signaling pathway leads to initiation of the Janus Kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) pathway that contributes to the pathogenesis of GvHD. The key role of JAK signaling in normal and abnormal lymphocyte development and function, along with the cytotoxic effects of its inhibitor INCB18424 (Ruxolitinib) on leukemia cells, prompted us to hypothesize that this selective JAK1 and 2 inhibitor could be useful as anti-GvHD agent while maintaining antitumor activity. Since CP-690550, a more selective JAK3 inhibitor, was recently shown to protect against GvHD in mouse models, we also tested whether blocking the JAK1/JAK2 pathway could be more effective in preventing GvHD. Methods: To assess the therapeutic effect of pharmacologic modulation of JAK1 and 2 on GvHD, a major histocompatibility complex (MHC) mismatched HSCT mouse model was used. Recipient BALB/c mice were lethally irradiated and treated either with spleen and bone marrow (BM) cells from C57BL/6 (B6) donors (GvHD cohort, n=8), or with spleen and BM cells from B6 donors along with INCB18424 90mg/kg/day at days -1 to 13 (INCB18424 cohort, n=10) or with CP-690550 15mg/kg/day (CP-690550 cohort, n=8) at days -1 to 13. Syngeneic transplants (B6-B6, n=6) and BALB/c recipients treated with B6 BM cells only (control cohort, n=8) were also included as controls. Mice were characterized for GvHD by monitoring overall survival and weight loss. Recipient mice were sacrificed and tissues harvested on day 14 and 30 post transplant and GvHD confirmed by histology. Results: All mice in the GvHD cohort had clinical evidence of GvHD (weight loss, generalized erythema of the skin and poor fur quality) by day 14. The INCB18424 treated mice showed markedly reduced weight loss along the time of observation when compared to the GvHD cohort. Animals in the CP-690550 cohort tended to gain weight during the time of treatment (day-1 to 13), but thereafter they exhibited reduced body weight similar to that observed in the GvHD cohort. The histological examination of the stomach, liver, skin and intestine obtained at day 14 revealed no sign of GvHD in the control group as well as in the INCB18424 group. On the other hand, mild to moderate signs of GvHD were present in the tissues of CP-690550 treated mice and extensive inflammation and disruption of the normal architecture of the tissues was observed in the GvHD group. To determine whether INCB18424 treatment affected alloreactive CD4+ T cells, total spleen T cells were harvested at day 14 from the GvHD cohort and from recipients either of INCB18424 or CP-690550. Total spleen T cells were co-cultured with BM derived BALB/c (recipient-derived) or C57BL/6 (donor-derived) dendritic cells (DCs). After 24h, T cells alloreactivity was determined by IFN-γ production assessed by intracellular staining. As expected, T cells from GvHD mice showed significantly higher alloreactivity against BALB/c DCs compared to the reactivity observed against syngeneic B6 DCs (5.24% and 0.84% respectively, p Conclusions: The inhibition of Jak/STAT signaling using the sensitive and specific inhibitor of Jak1/Jak2, INCB18424, conferred effective protection from aGvHD in a HSCT mouse model. INCB18424 treatment was more effective than the targeting of JAK3 with CP-690550. In fact, CP-690550 administered during GvHD induction was not completely sufficient to restore the normal weight and to prevent the histological appearance of GvHD whereas INCB18424 was. INCB18424 protected mice against acute GvHD by significantly decreasing alloreactive CD4 T cells. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
19. G-CSF Stem Cell Mobilization Is Protective Against Acute Graft-Versus-Host Disease (aGvHD) by Increasing Myeloid-Derived Suppressor Cell (MDSC) Frequencies
- Author
-
Fabiana Riva, Jacopo Mariotti, Cristiana Carniti, Antonio Vendramin, Silvia Gimondi, Anisa Bermema, and Paolo Corradini
- Subjects
business.industry ,Donor selection ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Filgrastim ,Total body irradiation ,Biochemistry ,Granulocyte macrophage colony-stimulating factor ,medicine.anatomical_structure ,medicine ,Myeloid-derived Suppressor Cell ,IL-2 receptor ,Bone marrow ,business ,medicine.drug - Abstract
Abstract 1898 Background: In mice, graft-versus-host disease (GvHD) can be abrogated by ex vivo expanded, bone marrow derived myeloid-derived suppressor cells (MDSCs) generated in the presence of GM-CSF, G-CSF and IL-13 (Highfill et al). Whether MDSCs play a role in human allogeneic hematopoietic stem cell transplantation (allo-HSCT) is still unclear. We hypothesized that G-CSF stem cell mobilization may be protective from acute GvHD (aGvHD) by increasing MDSC frequencies. Methods: G-CSF-mobilized peripheral blood (PB) samples were collected from 40 healthy unrelated donors (median age 34, range 20–43, male/female 31/9) who received G-CSF (Filgrastim) at 10 μg/kg/day for 5 days. Donor selection had been performed according to standard criteria, including molecular typing for HLA-A,-B,-C, DRB1, and DQB1. Donors were 10/10 HLA-matched (MUD) in 20 cases, 9/10 in 13 cases and 8/10 in the remaining 7 cases (MMUD). Patients (median age 46, range 18–67) received reduced intensity conditioning based or low-dose total body irradiation (TBI 2Gy) (8), Fludarabine-Rabbit Antithymocyte Globulin (ATG) (9) or Thiotepa-ATG (23). Diagnosis were lymphomas (29), myelomas (8), acute myeloid leukemia (3). GvHD prophylaxis was cyclosporine plus either methotrexate (36) or mycophenolate mofetil (4). As controls, PB samples were collected from 10 healthy adults. Informed consent was obtained from all subjects. Cells were characterized using flow cytometry with Abs against CD3;CD14;CD16;CD19;CD20;CD56;CD11b;HLADR;CD33. The frequencies of MDSCs and T regulatory cells (Tregs, CD4+CD25+CD127-FoxP3+) in the grafts were correlated with the clinical characteristics and outcome of the 40 patients. To verify that G-CSF treatment increases the number of MDSCs that are transferred with the graft and prevent GvHD, a major histocompatibility complex (MHC) mismatched HSCT mouse model was also used. After lethal irradiation, recipient BALB/c mice received spleen and bone marrow (BM) cells from C57BL/6 (B6) donors (GvHD cohort, n=5) or BM cells only (negative control, n=3). To generate MDSC enriched allografts, donor mice were treated with G-CSF (5 μg/d for 5 days) and thereafter BM cells and spleen cells were transferred in recipient mice (MDSC cohort, n=5). Results: Expansion of MDSCs (Lin-/LoHLADR−CD33+CD11b+) in the PB of G-CSF-treated unrelated donors was found with respect to steady state control individuals (p< 0.03, Mann Whitney-U). Acute GvHD occurred in 16 of 40 patients (40%). There was no significant correlation between the incidence of aGvHD and the degree of HLA incompatibility or the presence of donor-recipient sex mismatches. Neither the conditioning regimens nor the GvHD prophylaxis had effect on risk of aGvHD. There was no correlation between the number of Tregs infused and the occurrence of GvHD.Conversely, aGvHD patients received grafts containing significantly lower number of MDSCs when compared to non- aGvHD patients (p < 0.006, Mann Whitney-U). The ability of MDSC levels in the graft to predict the occurrence of aGvHD was determined by the receiver operating characteristic (ROC) curve: sensitivity was 100%, specificity 60%, AUC=0.768. The immunosuppressive activity of the MDSCs on activated T lymphocytes was confirmed in vitro. To confirm these results, we set up a MHC mismatched HSCT mouse model. G-CSF treatment induced a significant increase in MDSCs (up to four fold, p Conclusions: G-CSF mobilization significantly increases circulating MDSC both in Matched Unrelated Donors and in mice. In humans, a significant correlation between the frequencies of MDSC present in the graft and the incidence of aGvHD was found. This result was further confirmed using a MHC mismatched HSCT mouse model. Mice treated with G-CSF received grafts containing higher numbers of MDSCs and developed less severe aGvHD than the untreated mice. Taken together, these findings strongly suggest that G-CSF stem cell mobilization is protective against aGvHD through MDSCs augmentation and might be relevant to modify GvHD prophylaxis. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
20. Noninvasive Prediction of Acute Graft-Verus-Host Disease Following Allogeneic Hematopoietic Stem Cell Transplantation by Circulating miRNA Profiling
- Author
-
Antonio Vendramin, Silvia Gimondi, Cristiana Carniti, Mara Morelli, Anisa Bermema, Chiara Formica, Davide Lucini, Paolo Corradini, and Jacopo Mariotti
- Subjects
biology ,business.industry ,medicine.medical_treatment ,Immunology ,Spleen ,Cell Biology ,Hematology ,Human leukocyte antigen ,Hematopoietic stem cell transplantation ,medicine.disease ,Major histocompatibility complex ,Biochemistry ,Transplantation ,surgical procedures, operative ,medicine.anatomical_structure ,Immune system ,Graft-versus-host disease ,medicine ,biology.protein ,Bone marrow ,business - Abstract
Abstract 311 Background: Acute graft-versus-host disease (aGVHD) results in significant morbidity and mortality and remains the main complication of alloHSCT. Noninvasive, diagnostic and prognostic tests for aGVHD are currently lacking but essential to predict GVHD and to improve the safety and accessibility of alloHSCT. We hypothesized that the prospective analysis of miRNA expression profile in the plasma of allografted patients could allow for the detection of specific miRNAs with predictive role for aGVHD. Methods: After informed consent, we collected plasma samples from 10 healthy donors and 22 patients (median age: 59 and 41 years) who received unmanipulated alloHSCT (18 from Matched Unrelated Donors and 4 from HLA-matched siblings). Blood samples were collected weekly after HSCT and patients were monitored to assess aGVHD onset. MicroRNAs were isolated from the plasma and the miRNA expression profile examined using a quantitative PCR-method (TaqMan® Human microRNA Cards, Applied Biosystems). The results obtained were subsequently validated with specific miRNA Single Assays (Applied Biosystems). To verify whether the miRNAs emerged from the human studies represent markers of aGVHD and provide information regarding the involvement of specific target organs, a major histocompatibility complex (MHC) mismatched HSCT mouse model was used. Recipient BALB/c mice were lethally irradiated and treated either with spleen and bone marrow (BM) cells from C57BL/6 (B6) donors (GVHD cohort, n=22) or with BM cells only (negative control, n=18). Syngeneic transplants (B6àB6, n=6 were also included. Mice were characterized for GVHD onset by monitoring overall survival and weight loss. Recipient mice were sacrificed and tissues harvested on day 9, 14 and 18 post transplant and GVHD confirmed by histology and scored according to Foley et al, 2008. MiRNAs expression profile have been characterized in the plasma, skin, liver, colon and lymphocytes of GVHD and non-GVHD control cohorts Results: Three of 22 patients developed intestinal GVHD (grade 2) and 9 of 22 patients developed cutaneous GVHD (grade 2–3). By comparing the circulating miRNAs expression profiles of GVHD patients and non GVHD patients, we identified a group of 8 differentially expressed miRNAs (miR-136, 194, 203, 367, 148b, 196b, 26a, 340) (p Conclusions: Considering the noninvasive characteristics of plasma sampling and the reproducible and easy detection of miRNAs, our results indicate that circulating miRNAs might represent a promising tool for the early diagnosis of aGVHD thus enhancing therapeutic success and increasing life expectancy of allografted patients. In addition the miRNA profiling of the target organs and lymphocytes of GVHD mice, allowed the identification of several deregulated genes that might play a role in the modulation of aGVHD and warrant further investigations. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2011
21. High-Dose Rituximab in the Conditioning Regimen Before Allogeneic Stem Cell Transplantation Reduces the Incidence of Acute Gvhd in B-Cell Lymphomas
- Author
-
Cristiana Carniti, Alessandro Levis, Paolo Corradini, Alberto Bosi, Cecilia Olivares, Angelo Michele Carella, Antonio Vendramin, Paolo Bartolomeo, Michele Falda, Silvia Gimondi, Francesca Patriarca, Barbara Sarina, and Anna Dodero
- Subjects
CD20 ,medicine.medical_specialty ,biology ,Cyclophosphamide ,business.industry ,Immunology ,Follicular lymphoma ,Cell Biology ,Hematology ,ThioTEPA ,medicine.disease ,Biochemistry ,Gastroenterology ,Fludarabine ,Surgery ,Transplantation ,Internal medicine ,medicine ,biology.protein ,Cumulative incidence ,Rituximab ,business ,medicine.drug - Abstract
Abstract 4546 Background: Reduced-intensity conditioning (RIC) followed by allogeneic stem cell transplantation (alloSCT) is an effective salvage therapy for relapsed lymphomas. The present GITMO study is a prospective multicenter phase II trial for patients affected by relapsed CD20 positive lymphomas. Compared with the previous thiotepa/fludarabine/cyclophosphamide GITMO protocol (Leukemia 2007), the thiotepa dose is increased, and high-dose Rituximab is included in the regimen to improve the outcome and possibly modulate the incidence of acute GVHD. Aims: Primary end-point was 1-year progression-free survival; secondary endpoints were non-relapse mortality and incidence of acute and chronic GVHD. Methods: Fifty-seven patients (pts) were enrolled so far in the study and 49 are evaluable for analysis. Treatment plan consisted of high-dose R (500 mg/ms on day -6) followed by thiotepa (12 mg/kg), fludarabine (60 mg/kg) and cyclophosphamide (60 mg/kg). Graft-versus-host disease (GVHD) prophylaxis included cyclosporine and mini-methotrexate; ATG (7.5 mk/kg) was only added for pts allografted from one antigen mismatched sibling or unrelated donors. Histopathological subtypes included 24 aggressive (HG) (n= 17 diffuse large B-cell lymphomas, n= 7 mantle cell lymphomas) and 25 indolent lymphomas (LG) (n= 13 follicular lymphomas, n= 12 small lymphocytic/chronic lymphocytic leukemia). Patients were allografted from related siblings (SIB) (n= 32 matched, n=1 one single mismatched) or unrelated donors (UD) (n=11 matched, n=5 mismatched). All the pts had chemosensitive disease (n=20, 41% in complete remission) and 26 (53%) came from a failed autoSCT. Results: At a median follow-up of 13 months (range, 5–44 months), 36 pts are alive [n=27 (75%) in CR] and 13 died from any cause [n=6 for non-relapse mortality (NRM), n=7 for disease progression]. All the patients engrafted (94% had full donor chimerism at 3 months). The cumulative incidence (CI) of NRM was 13% at 1 year: 9% vs 19% for SIB and MUD (p=0.3), and 9% versus 16% for for LG and HG (p=0.3), respectively. In total only 11 of 49 pts had acute GVHD (n=8 grade II, n=3 grade III) with an estimated CI of 21% at 100 days. In the previous GITMO study the incidence was 35% with SIB only. Forty pts are evaluable for chronic GVHD with an estimated CI of 41% and 47% at 1 and 2 year, respectively (n=11 limited, n=3 extensive). Infections after engraftment requiring hospitalization or intravenous treatment were evaluable in 46 pts (n=3 excluded for early death). The overall incidence of infections was 58% (n=27) including 5 pts experienced sepsis and 10 pts pneumonia. Preliminary data on immune-reconstitution at 1 year showed: 1) low number of circulating B cells (median CD19+/ul: 129/ul) with an expansion of naive cells (IgD+, CD27-); 2) the median value of IgM was 89 mg/dl whereas IgG and IgA remained at low levels. The CI of relapse was 26% and 37% at 1 year and 2 years, respectively. In the indolent and aggressive groups, OS estimates at 2 years were 79% (95%CI, 52%-91%) and 61% (95 CI, 38%-77%) and PFS estimates were 53% (95%CI, 23%-76%) and 48% (95% CI, 27%-66%), respectively. Conclusions: The present data suggest that the administration of high-dose R is feasible and causes an unexpected reduction of the incidence of acute GVHD without increasing the NRM and the incidence of severe infections complications. Complete data evaluating the effects of R on immune reconstitution are ongoing. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2011
22. Analysis of Circulating Microrna Expression Profile In Patients Transplanted From Matched Unrelated Donors (MUD) Identifies mir203 as a Putative Marker of aGVHD
- Author
-
Cristiana Carniti, Paolo Corradini, Silvia Gimondi, Mara Morelli, Anisa Bermema, and Antonio Vendramin
- Subjects
medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Disease ,Hematopoietic stem cell transplantation ,Biology ,medicine.disease ,Biochemistry ,Pathogenesis ,Transplantation ,Circulating MicroRNA ,surgical procedures, operative ,Cytokine ,Graft-versus-host disease ,microRNA ,medicine - Abstract
Abstract 2308 Introduction: Allogeneic haemopoietic-stem-cell transplantation (HSCT) is the treatment of choice for many malignant and non-malignant disorders. Despite the recent advances in post-transplant immunosuppressive therapy, Graft-versus-Host Disease (GVHD) still represents the major life-threatening complication, developing in a substantial number of HSCT patients and resulting in poor outcome. Recent studies have indicated that microRNAs (miRNAs) circulate in a stable, cell-free form in the bloodstream and that the abundance of specific miRNAs in plasma or serum can serve as biomarkers of cancer and other diseases. We examined plasma microRNA (miRNA) expression levels from patients transplanted from matched unrelated donor (MUD) to assess their clinical application for diagnosing and monitoring GVHD. Methods: Having obtained an informed consent, we collected plasma samples from 11 patients who received unmanipulated HSCT from MUDs. After HSCT, 5 of 11 patients developed acute GVHD. Blood samples were collected serially at day +30, +60, +90, +150 after HSCT. MicroRNAs were isolated from the peripheral blood (PB) plasma using a modified mirVana™ miRNA Isolation Kit (Ambion Inc). The miRNAs expression profile was examined using a quantitative PCR-method (TaqMan ® Human microRNA cards, Applied Biosystems) that allows the analysis of 384 human miRNAs by low density array technology. Plasma samples of normal subjects have been included in the study. Relative quantification of miRNA expression was calculated with the 2-ΔΔCt method. The data were normalized respect to hsa-mir-122 and relative to a calibrator sample (average of normal subjects plasma samples). Results: Initial analysis showed that miRNAs are stable and detectable in all plasma samples from MUD transplanted patients. Among the 384 mirRNAs analyzed we identified a panel of mirRNAs that may have a predictive role for GVHD. Among these, we have identified mir203 which is upregulated in patients prior to the onset of GVHD and its expression decreases upon therapy. Mir203 acts downregulating SOCS3, the endogenous regulator of cytokine signaling through JAK-STAT3 which is involved in the activation of the immune response. The importance of SOCS3 in the pathogenesis of GVHD has been recenly demonstrated (Hill et al. Blood, 2010) confirming the potential regulatory role of mir203 in this disease. Of interest, the evaluation of mir203 expression levels at day +30 after HSCT is able to clearly classify the patients who will develop GVHD from those who will not (p-value Conclusions: Profiling of mir203 in a wider and heterogeneous group of patients receiving allo-HSCT is ongoing to extend and confirm the present findings. Although preliminary, these results indicate that the detection of circulating miRNAs might provide new complementary markers of GVHD. In particular the elevated plasma level of mir203 at day +30 after HSCT may be used to predict the risk of developing GVHD. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2010
23. Circulating miRNA panel for prediction of acute graft-versus-host disease in lymphoma patients undergoing matched unrelated hematopoietic stem cell transplantation
- Author
-
Silvia Gimondi, Paolo Corradini, Anisa Bermema, Cristiana Carniti, Giulia Biancon, Matteo Dugo, Alessandra Cavanè, and Antonio Vendramin
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,Time Factors ,Adolescent ,Lymphoma ,medicine.medical_treatment ,Gene Expression ,Graft vs Host Disease ,Disease ,Hematopoietic stem cell transplantation ,Pathogenesis ,03 medical and health sciences ,Young Adult ,immune system diseases ,hemic and lymphatic diseases ,microRNA ,Genetics ,medicine ,Cluster Analysis ,Humans ,Transplantation, Homologous ,Gene Regulatory Networks ,Young adult ,Molecular Biology ,Aged ,integumentary system ,business.industry ,Gene Expression Profiling ,Hematopoietic Stem Cell Transplantation ,Reproducibility of Results ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Prognosis ,Combined Modality Therapy ,Circulating MicroRNA ,MicroRNAs ,030104 developmental biology ,surgical procedures, operative ,Immunology ,Female ,business ,Biomarkers ,Blood sampling - Abstract
Acute graft-versus-host disease (aGVHD) results in significant morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Noninvasive diagnostic and prognostic tests for aGVHD are currently lacking, but would be beneficial in predicting aGVHD and improving the safety of allo-HSCT. Circulating microRNAs exhibit marked stability and may serve as biomarkers in several clinical settings. Here, we evaluated the use of circulating microRNAs as predictive biomarkers of aGVHD in lymphoma patients after allo-HSCT from matched unrelated donors (MUDs). After receiving informed consent, we prospectively collected plasma samples from 24 lymphoma patients before and after unmanipulated MUD allo-HSCT; microRNAs were then isolated. Fourteen patients developed aGVHD symptoms at a median of 48 days (range: 32–90) post-transplantation. Two patients developed intestinal GVHD, eight cutaneous GVHD, and four multiorgan GVHD. The microRNA expression profile was examined using quantitative real-time polymerase chain reaction (qRT-PCR). MicroRNAs 194 and 518f were significantly upregulated in aGVHD samples compared with samples taken from non-aGVHD patients. Remarkably, these upregulated microRNAs could be detected before the onset of aGVHD. Pathway prediction analysis indicated that these microRNAs may regulate critical pathways involved in aGVHD pathogenesis. Considering the noninvasive characteristics of plasma sampling and the feasibility of detecting miRNAs after allo-HSCT using real-time polymerase chain reaction, our results indicate that circulating microRNAs have the potential to enable an earlier aGVHD diagnosis and might assist in individualizing therapeutic strategies after MUD allo-HSCT. Nevertheless, standardization of blood sampling and analysis protocols is mandatory for the introduction of miRNA profiling into routine clinical use.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.