136 results on '"Evdoxia Hatjiharissi"'
Search Results
2. P835: IN SILICO PREDICTION REVEALS PUTATIVE T-CELL CLASS I/II NEOEPITOPES WITHIN THE CLONOTYPIC IMMUNOGLOBULIN HEAVY AND LIGHT CHAINS IN PATIENTS WITH MULTIPLE MYELOMA
- Author
-
Glykeria Gkoliou, Nikos Pechlivanis, Sofia Chatzileontiadou, Charikleia Xydopoulou, Christina Frouzaki, Georgios Karakatsoulis, Elisavet Vlachonikola, Marina Gerousi, Fotis Psomopoulos, Alexandra Siorenta, Maria Papaioannou, Katerina Chlichlia, Kostas Stamatopoulos, Evdoxia Hatjiharissi, and Anastasia Chatzidimitriou
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2023
- Full Text
- View/download PDF
3. Differences in the immunoglobulin gene repertoires of IgG versus IgA multiple myeloma allude to distinct immunopathogenetic trajectories
- Author
-
Glykeria Gkoliou, Andreas Agathangelidis, Georgos Karakatsoulis, Chrysavgi Lalayanni, Apostolia Papalexandri, Alejandro Medina, Elisa Genuardi, Katerina Chlichlia, Evdoxia Hatjiharissi, Maria Papaioannou, Evangelos Terpos, Cristina Jimenez, Ioanna Sakellari, Simone Ferrero, Marco Ladetto, Ramon Garcia Sanz, Chrysoula Belessi, and Kostas Stamatopoulos
- Subjects
multiple myeloma ,immunogenetics ,immunoglobulin isotypes ,immunoglobulin a ,immunoglobulin g ,immunoglobulin gene repertoire ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The analysis of the immunogenetic background of multiple myeloma (MM) has proven key to understanding disease ontogeny. However, limited information is available regarding the immunoglobulin (IG) gene repertoire in MM cases carrying different heavy chain isotypes. Here, we studied the IG gene repertoire in a series of 523 MM patients, of whom 165 and 358 belonged to the IgA and IgG MM groups, respectively. IGHV3 subgroup genes predominated in both groups. However, at the individual gene level, significant (p
- Published
- 2023
- Full Text
- View/download PDF
4. Chromatin-based, in cis and in trans regulatory rewiring underpins distinct oncogenic transcriptomes in multiple myeloma
- Author
-
Jaime Alvarez-Benayas, Nikolaos Trasanidis, Alexia Katsarou, Kanagaraju Ponnusamy, Aristeidis Chaidos, Philippa C. May, Xiaolin Xiao, Marco Bua, Maria Atta, Irene A. G. Roberts, Holger W. Auner, Evdoxia Hatjiharissi, Maria Papaioannou, Valentina S. Caputo, Ian M. Sudbery, and Anastasios Karadimitris
- Subjects
Science - Abstract
Despite extensive genetic heterogeneity, nearly half of all multiple myeloma (MM) cases are driven by cyclin D2 (CCND2) over-expression. Here the authors dissect the chromatin landscape of MM to provide insights into the transcriptional regulatory landscape driving MM and divergent transcriptomes corresponding to different MM genetic subtypes.
- Published
- 2021
- Full Text
- View/download PDF
5. Exploring the current molecular landscape and management of multiple myeloma patients with the t(11;14) translocation
- Author
-
Michael D. Diamantidis, Sofia Papadaki, and Evdoxia Hatjiharissi
- Subjects
translocation t(11 ,14) ,multiple myeloma ,precision medicine ,BCL-2 ,venetoclax ,genetic abnormalities ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Multiple myeloma (MM) is a genetically complex disease. The key myeloma-initiating genetic events are hyperdiploidy and translocations involving the immunoglobulin heavy chain (IgH) enhancer on chromosome 14, which leads to the activation of oncogenes (e.g., CCND1, CCND3, MAF, and MMSET). The t(11;14) translocation is the most common in MM (15%–20%) and results in cyclin D1 (CCND1) upregulation, which leads to kinase activation and tumor cell proliferation. Notably, t(11;14) occurs at a higher rate in patients with plasma cell leukemia (40%) and light chain amyloidosis (50%). Patients with myeloma who harbor the t(11;14) translocation have high levels of the anti-apoptotic protein B-cell lymphoma 2 (BCL2). Multiple studies demonstrated that the presence of t(11;14) was predictive of BCL2 dependency, suggesting that BCL2 could be a target in this subtype of myeloma. Venetoclax, an oral BCL2 inhibitor, has shown remarkable activity in treating relapsed/refractory MM patients with t(11;14) and BCL2 overexpression, either as monotherapy or in combination with other anti-myeloma agents. In this review, we describe the molecular defects associated with the t(11;14), bring into question the standard cytogenetic risk of myeloma patients harboring t(11;14), summarize current efficacy and safety data of targeted venetoclax-based therapies, and discuss the future of individualized or precision medicine for this unique myeloma subgroup, which will guide optimal treatment.
- Published
- 2022
- Full Text
- View/download PDF
6. Bing-Neel Syndrome: Real-Life Experience in Personalized Diagnostic Approach and Treatment
- Author
-
Dimitrios Kotsos, Sofia Chatzileontiadou, Athanasia Apsemidou, Anna Xanthopoulou, Aikaterini Rapi, Christina Frouzaki, and Evdoxia Hatjiharissi
- Subjects
Waldenström’s Macroglobulinemia ,Bing-Neel syndrome ,central nervous system infiltration ,ibrutinib ,real-life data ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The involvement of the central nervous system (CNS) in Waldenström’s Macroglobulinemia (WM) is a rare extramedullary manifestation of the disease known as Bing-Neel syndrome (BNS). To expand our understanding of this disease manifestation, we conducted a retrospective analysis of the incidence of BNS in 86 consecutive patients with WM [70% male, median age 65 years (range 33-86)] seen in our center during a 30-year period. Six patients (7%) from this group were diagnosed with BNS. The median period of time between WM diagnosis and BNS diagnosis was 6.8 years (range 2.3-15). They demonstrated a range of neurological deficits, including transient expressive aphasia, impaired vision, resting hand tremor, foot drop, and headache. Between the onset of symptoms and the diagnosis of BNS, the median time interval was 12.5 months (range 1-30). The diagnosis was made not on the basis of neurological symptoms or radiological evidence, but on the basis of the presence of WM cells in cerebrospinal fluid (CSF). Intrathecal chemotherapy with methotrexate, cytarabine, and dexamethasone (IT MTX, ARA-C, DEX) was used as front-line treatment, followed by intensive immunochemotherapy with rituximab, high-dose MTX, and ARA-C (R-Hi MTX/ARA-C) in three patients who were fit enough to receive this type of cytotoxic regimen, and rituximab plus bendamustine (R-Benda) in two patients who simultaneously required treatment for WM. Ibrutinib was administered to five patients (three as consolidation and two for initial treatment). All patients responded to front-line treatment, with four (67%) achieving partial response (PR) and two (33%) achieving complete response (CR). This study provides insight into the clinical presentation, diagnostic and treatment options, as well as the outcome of patients who have BNS.
- Published
- 2022
- Full Text
- View/download PDF
7. Competing risk survival analysis in patients with symptomatic Waldenström macroglobulinemia: the impact of disease unrelated mortality and of rituximab-based primary therapy
- Author
-
Efstathios Kastritis, Marie-Christine Kyrtsonis, Pierre Morel, Maria Gavriatopoulou, Evdoxia Hatjiharissi, Argirios S. Symeonidis, Amalia Vassou, Panagiotis Repousis, Sossana Delimpasi, Anastasia Sioni, Evrydiki Michalis, Michail Michael, Elina Vervessou, Michael Voulgarelis, Costantinos Tsatalas, Evangelos Terpos, and Meletios A. Dimopoulos
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2015
- Full Text
- View/download PDF
8. MAF functions as a pioneer transcription factor that initiates and sustains myelomagenesis
- Author
-
Alexia Katsarou, Nikolaos Trasanidis, Kanagaraju Ponnusamy, Ioannis V Kostopoulos, Jaime Alvarez-Benayas, Foteini Papaleonidopoulou, Keren Keren, Pierangela MR Sabbattini, Niklas Feldhahn, Maria Papaioannou, Evdoxia Hatjiharissi, Ian M Sudbery, Aristeidis Chaidos, Valentina S Caputo, and Anastasios Karadimitris
- Subjects
Hematology - Abstract
Deregulated expression of lineage-affiliated transcription factors is a major mechanism of oncogenesis. However, how deregulation of non-lineage affiliated TF impacts chromatin to initiate oncogenic transcriptional programmes is not well known. To address this, we studied the chromatin effects imposed by oncogenic MAF as the cancer-initiating driver in the plasma cell cancer multiple myeloma. We found that the ectopically expressed MAF endows myeloma plasma cells with migratory and proliferative transcriptional potential. This potential is regulated by activation of enhancers and super-enhancers, previously inactive in normal B cells and plasma cells, and in co-operation of MAF with the plasma cell-defining TF IRF4. Forced ectopic MAF expression confirms the de novo ability of oncogenic MAF to convert transcriptionally inert chromatin to active chromatin with features of super-enhancers, leading to activation of the MAF-specific oncogenic transcriptome and acquisition of cancer-related cellular phenotypes such as CCR1-dependent cell migration. These findings establish oncogenic MAF as a pioneer transcription factor that can initiate as well as sustain oncogenic transcriptomes and cancer phenotypes. However, despite its pioneer function, myeloma cells remain MAF-dependent thus validating oncogenic MAF as a therapeutic target that would be able to circumvent the challenges of subsequent genetic diversification driving disease relapse and drug resistance.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure S2 from The Stat3/5 Signaling Biosignature in Hematopoietic Stem/Progenitor Cells Predicts Response and Outcome in Myelodysplastic Syndrome Patients Treated with Azacitidine
- Author
-
Ioannis Kotsianidis, Constantinos Tsatalas, Andreas Scorilas, Vassiliki Pappa, Emmanuil Spanoudakis, Helen A. Papadaki, Evdoxia Hatjiharissi, Maria Papaioannou, Vassilia Garypidou, Sofia Vakalopoulou, Evangelia Nakou, Panagiotis G. Adamopoulos, Christos K. Kontos, Athanasios G. Galanopoulos, Sotirios G. Papageorgiou, Theodoros P. Vassilakopoulos, Eleftheria Lamprianidou, and Paraskevi Miltiades
- Abstract
Significantly higher basal levels of Stat3 in patients with poor-risk cytogenetics.
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Table S1 from The Stat3/5 Signaling Biosignature in Hematopoietic Stem/Progenitor Cells Predicts Response and Outcome in Myelodysplastic Syndrome Patients Treated with Azacitidine
- Author
-
Ioannis Kotsianidis, Constantinos Tsatalas, Andreas Scorilas, Vassiliki Pappa, Emmanuil Spanoudakis, Helen A. Papadaki, Evdoxia Hatjiharissi, Maria Papaioannou, Vassilia Garypidou, Sofia Vakalopoulou, Evangelia Nakou, Panagiotis G. Adamopoulos, Christos K. Kontos, Athanasios G. Galanopoulos, Sotirios G. Papageorgiou, Theodoros P. Vassilakopoulos, Eleftheria Lamprianidou, and Paraskevi Miltiades
- Abstract
Multivariate analysis of prognostic factors for overall survival in the patient cohort (n=74)
- Published
- 2023
- Full Text
- View/download PDF
11. Data from The Stat3/5 Signaling Biosignature in Hematopoietic Stem/Progenitor Cells Predicts Response and Outcome in Myelodysplastic Syndrome Patients Treated with Azacitidine
- Author
-
Ioannis Kotsianidis, Constantinos Tsatalas, Andreas Scorilas, Vassiliki Pappa, Emmanuil Spanoudakis, Helen A. Papadaki, Evdoxia Hatjiharissi, Maria Papaioannou, Vassilia Garypidou, Sofia Vakalopoulou, Evangelia Nakou, Panagiotis G. Adamopoulos, Christos K. Kontos, Athanasios G. Galanopoulos, Sotirios G. Papageorgiou, Theodoros P. Vassilakopoulos, Eleftheria Lamprianidou, and Paraskevi Miltiades
- Abstract
Purpose: Azacitidine is the mainstay of high-risk myelodysplastic syndromes (MDS) therapy, but molecular predictors of response and the mechanisms of resistance to azacitidine remain largely unidentified. Deregulation of signaling via Stat3 and Stat5 in acute myeloid leukemia (AML) is associated with aggressive disease. Numerous genes involved in cell signaling are aberrantly methylated in MDS, yet the alterations and the effect of azacitidine treatment on Stat3/5 signaling in high-risk MDS have not been explored.Experimental Design: We assessed longitudinally constitutive and ligand-induced phospho-Stat3/5 signaling responses by multiparametric flow cytometry in 74 patients with MDS and low blast count AML undergoing azacitidine therapy. Pretreatment Stat3/5 signaling profiles in CD34+ cells were grouped by unsupervised clustering. The differentiation stage and the molecular properties of the CD34+ G-CSF–inducible Stat3/5 double-positive subpopulation were performed by flow cytometry and quantitative real-time PCR in isolated MDS progenitors.Results: The pretreatment Stat3/5 signaling profiles in CD34+ cells correlated strongly with response and cytogenetics and independently predicted event-free survival. We further identified a CD34+ G-CSF–inducible Stat3/5 double-positive subpopulation (DP subset) whose pretreatment levels were inversely associated with treatment response and cytogenetics. The kinetics of the DP subset followed the response to azacitidine and the disease course, whereas its molecular characteristics and cellular hierarchy were consistent with a leukemia propagating cell phenotype.Conclusions: Our findings provide a novel link among Stat3/5 signaling and MDS pathobiology and suggest that the Stat3/5 signaling biosignature may serve as both a response biomarker and treatment target. Clin Cancer Res; 22(8); 1958–68. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Methods from The Stat3/5 Signaling Biosignature in Hematopoietic Stem/Progenitor Cells Predicts Response and Outcome in Myelodysplastic Syndrome Patients Treated with Azacitidine
- Author
-
Ioannis Kotsianidis, Constantinos Tsatalas, Andreas Scorilas, Vassiliki Pappa, Emmanuil Spanoudakis, Helen A. Papadaki, Evdoxia Hatjiharissi, Maria Papaioannou, Vassilia Garypidou, Sofia Vakalopoulou, Evangelia Nakou, Panagiotis G. Adamopoulos, Christos K. Kontos, Athanasios G. Galanopoulos, Sotirios G. Papageorgiou, Theodoros P. Vassilakopoulos, Eleftheria Lamprianidou, and Paraskevi Miltiades
- Abstract
Mutations analysis of the TET2 and TP53 coding regions using Next-Generation Sequencing (NGS)
- Published
- 2023
- Full Text
- View/download PDF
13. A novel quantitative method for assessing the therapeutic response to Tafamidis therapy in patients with cardiac TTR amyloidosis. A preliminary report
- Author
-
Argiris, Doumas, Thomas, Zegkos, Despoina, Parcharidou, Thomas, Gossios, Dimitris, Ntelios, Sofia, Chatzileontiadou, Emmanouil, Papanastasiou, Evdoxia, Hatjiharissi, Ioannis, Iakovou, and Georgios K, Efthimiadis
- Subjects
Male ,Amyloid Neuropathies, Familial ,Benzoxazoles ,Humans ,Technetium ,Radionuclide Imaging ,Aged - Abstract
Cardiomyopathy is a common manifestation of transthyretin amyloidosis (ATTR), leading to heart failure, associated with high morbidity and mortality. The aim of this study was to investigate the effect of Tafamidis treatment by means of cardiac radiotracer uptake on myocardial scintigraphy.Five male patients, mean age 76.2 years, with wild-type ATTR were included in the protocol. Total body scanning using technetium-99m-3,3-diphosphono-1,2-propanodicarboxylic acid (Heart-to-thigh ratio was improved (decreased) in four of the patients receiving Tafamidis, in keeping with lower uptake to the cardiac region. These patients also demonstrated a relatively favorable clinical response to Tafamidis. The patient evaluated bySequential HtT ratio measurements could potentially identify patients with a favorable response to Tafamidis treatment at earlier stages, compared to other imaging modalities or serological biomarkers.
- Published
- 2022
14. Daratumumab Improves Bone Turnover in Relapsed/Refractory Multiple Myeloma; Phase 2 Study “REBUILD”
- Author
-
Dimopoulos, Evangelos Terpos, Ioannis Ntanasis-Stathopoulos, Efstathios Kastritis, Evdoxia Hatjiharissi, Eirini Katodritou, Evangelos Eleutherakis-Papaiakovou, Evgenia Verrou, Maria Gavriatopoulou, Alexandros Leonidakis, Kyriaki Manousou, Sosana Delimpasi, Panagiotis Malandrakis, Marie-Christine Kyrtsonis, Maria Papaioannou, Argiris Symeonidis, and Meletios-Athanasios
- Subjects
multiple myeloma ,daratumumab ,bone metabolism ,biomarker ,clinical trial - Abstract
Biomarkers of bone turnover in serum are suggestive of bone dynamics during treatment in multiple myeloma (MM). We evaluated the role of daratumumab on bone remodeling among patients with relapsed/refractory MM in the prospective, open-label, phase 2 study REBUILD. Daratumumab was administered according to the approved indication. A total of 33 out of 57 enrolled patients completed 4 months of treatment. The median percent change from baseline to 4 months in C-terminal cross-linking telopeptide of type 1 collagen (CTX) (primary endpoint) was 3.9%, with 13 (39.4%) and 11 (33.3%) patients showing at least 20% and 30% reduction in CTX levels, respectively. The median percent decrease from baseline to 4 months in tartrate resistant acid phosphatase 5b (TRACP-5b) levels (co-primary endpoint) was 2.6%, with 10 (30.3%) and 6 (18.2%) patients showing at least 20% and 30% reduction in TRACP-5b levels, respectively. However, the changes in these markers of bone catabolism were not statistically significant. Furthermore, the levels of osteocalcin, bone-specific alkaline phosphatase and procollagen type-I N-pro-peptide (bone formation markers) increased from baseline to 4 months (secondary endpoints) by 18.4%, 92.6% and 10.2%, respectively. Furthermore, the median levels of dickkopf-1 and C-C motif ligand-3 showed a significant decrease at 4 months by 17.5% and 16.0%, respectively. In conclusion, daratumumab improved bone turnover by inducing bone formation and reducing osteoblast inhibition.
- Published
- 2022
- Full Text
- View/download PDF
15. Real-life Experience With Rituximab-CHOP Every 21 or 14 Days in Primary Mediastinal Large B-cell Lymphoma
- Author
-
STAMATIS J. KARAKATSANIS, MARIA BOUZANI, ARGYRIS SYMEONIDIS, MARIA K. ANGELOPOULOU, SOTIRIOS G. PAPAGEORGIOU, MICHAIL MICHAIL, GABRIELLA GAINARU, GEORGIA KOURTI, SOTIRIOS SACHANAS, CHRISTINA KALPADAKIS, EIRINI KATODRITOU, THEONI LEONIDOPOULOU, IOANNIS KOTSIANIDIS, ELEFTHERIA HATZIMICHAEL, MARIA KOTSOPOULOU, MARIA DIMOU, ELENI VARIAMIS, DIMITRIOS BOUTSIS, NICK KANELLIAS, MARIA N. DIMOPOULOU, EVRIDIKI MICHALI, GEORGE KARIANAKIS, PANTELIS TSIRKINIDIS, CHRYSSA VADIKOLIA, CHRISTOS POZIOPOULOS, ANNA PIGADITOU, EFFIMIA VRAKIDOU, THEOPHANIS ECONOMOPOULOS, LYDIA KYRIAZOPOULOU, MARINA P. SIAKANTARIS, MARIE-CHRISTINE KYRTSONIS, KONSTANTINOS ANARGYROU, MARIA PAPAIOANNOU, EVDOXIA HATJIHARISSI, ELISSAVET VERVESSOU, MARIA TSIROGIANNI, MARIA PALASSOPOULOU, EKATERINI STEFANOUDAKI, PANAYIOTIS ZIKOS, PANAYIOTIS TSIRIGOTIS, GERASSIMOS TSOUROUFLIS, THEODORA ASSIMAKOPOULOU, EVGENIA VERROU, HELEN PAPADAKI, POLIXENI LAMPROPOULOU, MELETIOS-ATHANASIOS DIMOPOULOS, VASSILIKI PAPPA, KOSTAS KONSTANTOPOULOS, THEMIS KARMIRIS, PARASKEVI ROUSSOU, PANAYIOTIS PANAYIOTIDIS, GERASSIMOS A. PANGALIS, and THEODOROS P. VASSILAKOPOULOS
- Subjects
Pharmacology ,Cancer Research ,Lymphoma, B-Cell ,General Biochemistry, Genetics and Molecular Biology ,immune system diseases ,Doxorubicin ,Vincristine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Prednisone ,Prospective Studies ,Rituximab ,Cyclophosphamide ,Research Article ,Retrospective Studies - Abstract
Background/Aim: Primary mediastinal large B-cell lymphoma (PMLBCL) is an aggressive B-cell non-Hodgkin lymphoma (NHL), whose prognosis has greatly improved since the incorporation of the anti-CD20 monoclonal antibody rituximab into current therapeutic regimens. Evidence, however, on the optimal time interval between consecutive chemoimmunotherapy (CIT) cycles is still scarce. This study aimed to evaluate the efficacy outcomes of the more commonly administered 3-weekly regimens to the biweekly ones in a PMLBCL patients’ population, who were mostly treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone every 21 days (R-CHOP-21) or R-CHOP-14. Patients and Methods: We retrospectively studied our cohort of consecutively treated PMLBCL patients, focusing on their treatment density, in order to determine possible differences in treatment outcomes. Results: CIT, in the form of both R-CHOP-21 as well as R-CHOP-14 (or similar regimens), is highly active in PMLBCL, with low rates of early treatment failure. In our cohort of patients, R-CHOP-14 did not result in a meaningful improvement of freedom from progression (FFP) or overall survival (OS). Conclusion: Both R-CHOP-14 and R-CHOP-21 are probably equally effective in PMLBCL, yet further, prospective, randomized studies are warranted to clarify whether dose-dense regimens can be associated with better disease control and long-term results.
- Published
- 2022
16. Identification a Novel Molecular Mechanism Underlying the Anti-CD38-Based Treatment Resistance in Multiple Myeloma Patients
- Author
-
Sophia Adamia, Daisuke Ogiya, Evdoxia Hatjiharissi, Michael P. Chu, Zuzana Chyra, Kenneth Wen, Yu-Tzu Tai, David M. Dorfman, Teru Hideshima, and Kenneth C. Anderson
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
17. T Cell Epitope Prediction within the Clonotypic Immunoglobulin Heavy and Light Chains in Patients with Multiple Myeloma
- Author
-
Glykeria Gkoliou, Nikos Pechlivanis, Sofia Chatzileontiadou, Chara Xydopoulou, Christina Frouzaki, Georgios Karakatsoulis, Elisavet Vlachonikola, Marina Gerousi, Fotis Psomopoulos, Maria Papaioannou, Katerina Chlichlia, Kostas Stamatopoulos, Evdoxia Hatjiharissi, and Anastasia Chatzidimitriou
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
18. Chromatin-based, in cis and in trans regulatory rewiring underpins distinct oncogenic transcriptomes in multiple myeloma
- Author
-
Ian Sudbery, Kanagaraju Ponnusamy, Aristeidis Chaidos, Holger W. Auner, Valentina S. Caputo, Philippa C. May, Alexia Katsarou, Marco Bua, Xiaolin Xiao, Maria Papaioannou, Anastasios Karadimitris, Jaime Alvarez-Benayas, Irene Roberts, Maria Atta, Evdoxia Hatjiharissi, and Nikolaos Trasanidis
- Subjects
Carcinogenesis ,Gene regulatory network ,General Physics and Astronomy ,Myeloma ,Gene regulatory networks ,Transcriptome ,hemic and lymphatic diseases ,ELEMENTS ,Cyclin D2 ,Cyclin D1 ,Multiple myeloma ,Multidisciplinary ,PROLIFERATION ,Chromatin ,Gene Expression Regulation, Neoplastic ,Multidisciplinary Sciences ,GENOME ,Enhancer Elements, Genetic ,Proto-Oncogene Proteins c-maf ,Science & Technology - Other Topics ,Systems biology ,Multiple Myeloma ,PROTEINS ,Science ,Plasma Cells ,Bone Marrow Cells ,Computational biology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,CLASSIFICATION ,Cell Line, Tumor ,medicine ,Humans ,Enhancer ,Gene ,Transcription factor ,Science & Technology ,Gene Expression Profiling ,General Chemistry ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Survival Analysis ,Repressor Proteins ,Case-Control Studies ,Ectopic expression - Abstract
Multiple myeloma is a genetically heterogeneous cancer of the bone marrow plasma cells (PC). Distinct myeloma transcriptome profiles are primarily driven by myeloma initiating events (MIE) and converge into a mutually exclusive overexpression of the CCND1 and CCND2 oncogenes. Here, with reference to their normal counterparts, we find that myeloma PC enhanced chromatin accessibility combined with paired transcriptome profiling can classify MIE-defined genetic subgroups. Across and within different MM genetic subgroups, we ascribe regulation of genes and pathways critical for myeloma biology to unique or shared, developmentally activated or de novo formed candidate enhancers. Such enhancers co-opt recruitment of existing transcription factors, which although not transcriptionally deregulated per se, organise aberrant gene regulatory networks that help identify myeloma cell dependencies with prognostic impact. Finally, we identify and validate the critical super-enhancer that regulates ectopic expression of CCND2 in a subset of patients with MM and in chronic lymphocytic leukemia., Despite extensive genetic heterogeneity, nearly half of all multiple myeloma (MM) cases are driven by cyclin D2 (CCND2) over-expression. Here the authors dissect the chromatin landscape of MM to provide insights into the transcriptional regulatory landscape driving MM and divergent transcriptomes corresponding to different MM genetic subtypes.
- Published
- 2021
19. Pomalidomide Plus Low Dose Dexamethasone in Relapsed/Refractory Multiple Myeloma Patients: Results of the Real-World 'Powerful' Study
- Author
-
Marie-Christine Kyrtsonis, Petros Patos, Evridiki Michali, Panagiotis Repousis, Argiris Symeonidis, Kiki Karvounis-Marolachakis, Christos Poziopoulos, Evdoxia Hatjiharissi, Gerassimos A. Pangalis, George Vassilopoulos, Eirini Katodritou, Helen A. Papadaki, Vassiliki Pappa, Panagiotis Zikos, Chrysavgi Lalayanni, Magda Dadakaridou, Theodora Assimakopoulou, Anastasia Pouli, Christos Georgopoulos, Evangelos Terpos, and Emmanouil Spanoudakis
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,education ,Immunology ,Low dose ,Cell Biology ,Hematology ,Pomalidomide ,medicine.disease ,Biochemistry ,Internal medicine ,Relapsed refractory ,medicine ,business ,health care economics and organizations ,Multiple myeloma ,Dexamethasone ,medicine.drug - Abstract
Introduction: Pomalidomide (POM) plus low-dose dexamethasone (POM/LoDex) is a standard of care for patients with relapsed/refractory multiple myeloma (RRMM), who have received ≥2 prior therapies, including lenalidomide (LEN) and bortezomib (BORT). In view of the limited real-world evidence on POM/LoDex effectiveness, this study aimed to provide insight into progression-free survival (PFS), response to treatment and drug utilization patterns in the routine clinical practice in Greece. Methods: 'POWERFUL' (NCT03353545) was a non-interventional, multicenter, retrospective and prospective study of adult RRMM patients, initiated on POM/LoDex between 01-Jan-2016 and 28-Feb-2019 per the approved label. Patients who had received up to one POM/LoDex cycle were consecutively enrolled between 16-Nov-2017 and 21-Feb-2019 (prospective recruitment phase). One month prior to completion of this phase, aiming to facilitate recruitment of the target size, patients, whose treatment with POM/LoDex was ongoing but >1 cycles had been received or for whom treatment was discontinued, were consecutively enrolled in reverse chronological order, based on POM/LoDex start date (retrospective recruitment). Patients were observed until the earliest point of disease progression (PD), death, informed consent (IC) withdrawal, treatment discontinuation for reason other than PD or start of next antimyeloma therapy, physician's decision, or last patient enrolled plus up to 12 months of treatment. IC or consent waiver for deceased subjects was obtained. Safety data were collected prospectively. Results: Eligible patients (N=99; 75 with prospective and 24 with a purely retrospective follow-up) were recruited by 18 hematology departments. The median (IQR) observation period was 8.8 (4.2-15.4) months. Baseline patient characteristics are presented in Table 1. Fifty patients (50.5%) started POM/LoDex as third-line treatment. POM was initiated at 4 mg/day on days 1-21 every 28 days in 75.8% of the patients. A median of 8 (range: 1-38) cycles were received at a median POM dose of 4 mg/day (range: 1-4), over 8.3 (range: 0.3-47.6) months. The POM dose reduction and interruption rates were 28.3% and 59.6%, respectively. Of the patients, 37.4% were treated for ≥ one year. Treatment was discontinued in 81.8%, due to PD (56.8%), safety reasons (22.2%), death (6.2%), other reasons (9.9%), and due to loss of follow-up (4.9%). The investigator-assessed overall response rate (≥partial response [PR]) was 32.3%; best response rates were: stringent complete or complete response (7.1%), very good PR (8.1%), PR (17.2%), minimal response (11.1%), stable disease (21.2%), PD (12.1%), and non-evaluable response (23.2%). Median time to response and duration of response in patients achieving ≥PR was 3.2 [95% confidence interval (CI): 2.6-3.6] and 15.8 (95%CI: 11.3-not reached) months, respectively. Over a Kaplan-Meier estimated median follow-up of 13.8 months, the median PFS was 10.5 months (95%CI: 7.4-14.4) [13.0 vs. 8.8 months for patients treated in the third vs. the fourth- and beyond line (log-rank p=0.494), and 13.0 vs. 8.8 months for patients treated with POM/LoDex only vs. those co-administered other antimyeloma agents (log-rank p=0.411)]. The estimated 6-, 12-, 24- and 36-month PFS rates were 70.3%, 48.3%, 20.1% and 12.0%, respectively. Multivariate Cox regression analysis of the impact of baseline characteristics on PFS indicated male sex [hazard ratio (HR)=2.08; 95%CI: 1.12-3.89; p=0.021] and higher than normal serum lactate dehydrogenase levels (HR=2.84; 95%CI: 1.39-5.81; p=0.004) as negative predictors of PFS. Over a median safety data collection period of 7.6 months (range: 0.4-18.6), the POM-related adverse events (ADR) incidence rate in the safety-evaluable population (i.e., 75 patients with prospective follow-up) was 42.7% (73 events). Only neutropenia (13.3%) was reported at a frequency ≥10%. The serious ADR rate was 18.7%, whereas grade ≥ 3 hematological and non-hematological ADR rates were 8.0%, and 5.3%, respectively. Conclusion: In this Real-World dataset from Greek centers, POM/LoDex, administered in the third-line and beyond setting of RRMM, displayed a longer PFS than in controlled clinical trials, which was not impacted by the treatment line and concurrent receipt of other antimyeloma agents. About one-third of patients achieved at least PR with an about 16-month response duration. Disclosures Terpos: Amgen: Honoraria, Research Funding; Sanofi: Honoraria; Celgene: Honoraria; Takeda: Honoraria, Other: travel expenses , Research Funding; Janssen: Honoraria, Research Funding; Genesis pharma SA: Honoraria, Other: travel expenses , Research Funding; BMS: Honoraria. Repousis:Genesis pharma SA: Membership on an entity's Board of Directors or advisory committees, Research Funding. Hatjiharissi:Roche: Honoraria; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Honoraria; Gilead: Membership on an entity's Board of Directors or advisory committees. Assimakopoulou:Genesis pharma SA: Research Funding. Vassilopoulos:Abbvie: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Research Funding. Pouli:Genesis pharma SA: Research Funding. Spanoudakis:Genesis pharma SA: Research Funding. Michali:Takeda: Research Funding; Genesis pharma SA: Research Funding. Pangalis:Genesis pharma SA: Research Funding. Poziopoulos:Genesis pharma SA: Research Funding. Kyrtsonis:Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Pappa:Genesis pharma SA: Research Funding. Symeonidis:Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GenesisPharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck Sharp & Dohme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; WinMedica: Research Funding; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Research Funding; Astellas: Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi/Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Georgopoulos:Genesis pharma SA: Research Funding. Zikos:Genesis pharma SA: Membership on an entity's Board of Directors or advisory committees, Research Funding. Papadaki:Genesis pharma SA: Membership on an entity's Board of Directors or advisory committees, Research Funding. Dadakaridou:Genesis pharma SA: Research Funding. Karvounis-Marolachakis:Genesis pharma SA: Current Employment. Patos:Genesis pharma SA: Current Employment. Katodritou:Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Theagenion Cancer Hospital: Current Employment; Takeda: Honoraria, Other: Expenses, Research Funding; Genesis Pharma: Honoraria, Other: Expenses, Research Funding; Abbvie: Research Funding; Karyopharm: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
20. A revised international prognostic score system for Waldenström’s macroglobulinemia
- Author
-
Marzia Varettoni, Bénédicte Hivert, Kamel Laribi, Alain Duhamel, Evangelos Terpos, Maria Gavriatopoulou, Efstathios Kastritis, Véronique Leblond, Amalia Vassou, Panagiotis Repousis, Marie-Christine Kyrtsonis, Euridyki Michalis, Michalis Michail, Eirini Katodritou, Argiris Symeonidis, Giampaolo Merlini, Evdoxia Hatjiharissi, Meletios A. Dimopoulos, Pierre Morel, Eric Durot, Loic Ysebaert, and Nikolaos Giannakoulas
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Proportional hazards model ,business.industry ,Clinical study design ,Macroglobulinemia ,Hematology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Chemoimmunotherapy ,030220 oncology & carcinogenesis ,Internal medicine ,Severity of illness ,medicine ,Rituximab ,business ,Prospective cohort study ,Survival rate ,medicine.drug - Abstract
A staging system was developed a decade ago for patients with Waldenstrom’s macroglobulinemia (WM), however, since then WM treatments have changed. A revised staging system could better capture prognosis of WM patients in the chemoimmunotherapy era. We developed a revised system based on data from 492 symptomatic patients with at least 3 years and a median of 7 years of follow up while an independent validation cohort included 229 symptomatic patients. We identified age (≤65 vs 66–75 vs ≥76 years), b2-microglobulin ≥ 4 mg/L, serum albumin 65 years and
- Published
- 2019
- Full Text
- View/download PDF
21. Primary gastric non-Hodgkin lymphomas: Recent advances regarding disease pathogenesis and treatment
- Author
-
Michael D. Diamantidis, Maria Papaioannou, and Evdoxia Hatjiharissi
- Subjects
Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,Mucosa-associated lymphoid tissue ,CHOP ,Immunochemotherapy ,Prednisone ,immune system diseases ,Stomach Neoplasms ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Retrospective Studies ,business.industry ,Gastroenterology ,Retrospective cohort study ,Minireviews ,General Medicine ,Lymphoma, B-Cell, Marginal Zone ,Diffuse large B-cell lymphoma ,medicine.disease ,Lymphoma ,Radiation therapy ,Primary gastric lymphoma ,Clinical research ,Rituximab-cyclophosphamide doxorubicin vincristine prednisone ,Lymphoma, Large B-Cell, Diffuse ,business ,Rituximab ,medicine.drug ,Extranodal non-Hodgkin’s lymphoma - Abstract
Primary gastric lymphomas (PGLs) are distinct lymphoproliferative neoplasms described as heterogeneous entities clinically and molecularly. Their main histological types are diffuse large B-cell lymphoma (DLBCL) or mucosa-associated lymphoma tissue. PGL has been one of the main fields of clinical research of our group in recent years. Although gastric DLBCLs are frequent, sufficient data to guide optimal care are scarce. Until today, a multidisciplinary approach has been applied, including chemotherapy, surgery, radiotherapy or a combination of these treatments. In this minireview article, we provide an overview of the clinical manifestations, diagnosis and staging of these diseases, along with their molecular pathogenesis and the most important related clinical published series. We then discuss the scientific gaps, perils and pitfalls that exist regarding the aforementioned studies, in parallel with the unmet need for future research and comment on the proper methodology for such retrospective studies. Aiming to fill this gap, we retrospectively evaluated the trends in clinical presentation, management and outcome among 165 patients with DLBCL PGL who were seen in our institutions in 1980-2014. The study cohort was divided into two subgroups, comparing the main 2 therapeutic options [cyclophosphamide doxorubicin vincristine prednisone (CHOP) vs rituximab-CHOP (R-CHOP)]. A better outcome with immunochemotherapy (R-CHOP) was observed. In the next 2 mo, we will present the update of our study with the same basic conclusion.
- Published
- 2021
22. Pomalidomide Plus Low-Dose Dexamethasone in Relapsed/Refractory Multiple Myeloma Patients: Results of the Real-World 'POWERFUL' Study
- Author
-
Eurydiki Michalis, Magdalini Dadakaridou, Argiris Symeonidis, Theodora Assimakopoulou, Vasiliki Pappa, Gerassimos A. Pangalis, Anastasia Pouli, Christos Georgopoulos, Chrysavgi Lalayanni, Panagiotis Repousis, Christos Poziopoulos, Evangelos Terpos, Panagiotis Zikos, Marie-Christine Kyrtsonis, Ioannis Ntanasis-Stathopoulos, Georgios Vassilopoulos, Maria Gavriatopoulou, Kiki Karvounis-Marolachakis, Helen A. Papadaki, Eirini Katodritou, Emmanouil Spanoudakis, and Evdoxia Hatjiharissi
- Subjects
medicine.medical_specialty ,animal structures ,ORR ,lenalidomide ,lcsh:Medicine ,pomalidomide ,Neutropenia ,Article ,PFS ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Prospective cohort study ,Adverse effect ,duration of response ,Multiple myeloma ,Lenalidomide ,Bortezomib ,business.industry ,lcsh:R ,General Medicine ,medicine.disease ,Pomalidomide ,multiple myeloma ,refractory ,030220 oncology & carcinogenesis ,Concomitant ,sense organs ,business ,030215 immunology ,medicine.drug - Abstract
The “POWERFUL” multicenter, retrospective, and prospective study investigated the effectiveness of pomalidomide plus low-dose dexamethasone (POM/LoDex) therapy in relapsed/refractory multiple myeloma in routine care in Greece. Ninety-nine eligible adult patients treated with POM/LoDex according to the approved label after having received ≥2 prior therapies, including lenalidomide and bortezomib, were consecutively enrolled between 16 November 2017 and 21 February 2019 in 18 hematology departments. Fifty patients (50.5%) started POM/LoDex as third-line treatment. During the treatment period (median: 8.3 months, range: 0.3–47.6 months), the median POM dose was 4 mg/day, and 31.3% of the patients received additional antimyeloma agents. The overall response rate was 32.3%. During a median follow-up period of 13.8 months (Kaplan–Meier estimate), the median progression-free survival (PFS) was 10.5 months (95% CI: 7.4–14.4). The PFS was not significantly different between patients receiving POM/LoDex in the third versus later line of therapy, nor between patients receiving concomitant antimyeloma therapy versus POM/LoDEx doublet. During the prospective safety data collection period (median: 7.6 months) among patients with prospective follow-up (N = 75), POM-related adverse event incidence rate was 42.7% (serious: 18.7%, grade ≥ 3 hematological POM-related adverse events: 8.0%). Only neutropenia (13.3%) was reported at a frequency ≥10%. In conclusion, in this real-world study, POM/LoDex displayed a long PFS with no new safety signals emerging.
- Published
- 2021
23. Identification of Very Low-Risk Subgroups of Patients with Primary Mediastinal Large B-Cell Lymphoma Treated with R-CHOP
- Author
-
Konstantinos Anargyrou, George Karianakis, Maria Kotsopoulou, Eleftheria Hatzimichael, Pavlina Konstantinidou, Maria Papaioannou, Chryssa Vadikolia, Evangelos Terpos, Katerina Megalakaki, Lydia Kyriazopoulou, Stamatios Karakatsanis, Anna Pigaditou, Theoni Leonidopoulou, Maria Dimou, Eleni Variamis, Michail Michail, Dimitrios Boutsis, Effimia Vrakidou, Gabriella Gainaru, Pantelis Tsirkinidis, Ioannis Kotsianidis, Kostas Konstantopoulos, Paraskevi Roussou, Maria N. Dimopoulou, Maria Palassopoulou, Theodora Assimakopoulou, Panayiotis Tsirigotis, Christina Kalpadakis, Maria K. Angelopoulou, Gerasimos Tsourouflis, Vassiliki Pappa, Evdoxia Hatjiharissi, Sotirios G. Papageorgiou, Theophanis Economopoulos, Themis Karmiris, Argyris Symeonidis, Meletios-Athanasios Dimopoulos, Christos Poziopoulos, Eirini Katodritou, Ekaterini Stefanoudaki, Panayiotis Zikos, Helen A. Papadaki, Marina P. Siakantaris, Theodoros P. Vassilakopoulos, G. Kourti, Maria Tsirogianni, Gerassimos A. Pangalis, Eurydiki Michalis, Panayiotis Panayiotidis, Sotirios Sachanas, Elissavet Vervessou, Marie-Christine Kyrtsonis, and Fotios Panitsas
- Subjects
Adult ,Cancer Research ,medicine.medical_specialty ,Multivariate analysis ,Hematologic Malignancies ,CHOP ,Gastroenterology ,Extranodal Disease ,03 medical and health sciences ,0302 clinical medicine ,International Prognostic Index ,Internal medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,Medicine ,Humans ,EPOCH (chemotherapy) ,Extranodal Involvement ,Cyclophosphamide ,business.industry ,medicine.disease ,Prognosis ,Lymphoma ,Oncology ,Doxorubicin ,Vincristine ,030220 oncology & carcinogenesis ,Prednisone ,Rituximab ,Lymphoma, Large B-Cell, Diffuse ,business ,030215 immunology ,medicine.drug - Abstract
Background R-CHOP can cure approximately 75% of patients with primary mediastinal large B-cell lymphoma (PMLBCL), but prognostic factors have not been sufficiently evaluated yet. R-da- EPOCH is potentially more effective but also more toxic than R-CHOP. Reliable prognostic classification is needed to guide treatment decisions. Materials and Methods We analyzed the impact of clinical prognostic factors on the outcome of 332 PMLBCL patients ≤65 years treated with R-CHOP ± radiotherapy in a multicenter setting in Greece and Cyprus. Results With a median follow-up of 69 months, 5-year freedom from progression (FFP) was 78% and 5-year lymphoma specific survival (LSS) was 89%. On multivariate analysis, extranodal involvement (E/IV) and lactate dehydrogenase (LDH) ≥2 times upper limit of normal (model A) were significantly associated with FFP; E/IV and bulky disease (model B) were associated with LSS. Both models performed better than the International Prognostic Index (IPI) and the age-adjusted IPI by Harrel's C rank parameter and Akaike information criterion. Both models A and B defined high-risk subgroups (13%–27% of patients [pts]) with approximately 19%–23% lymphoma-related mortality. They also defined subgroups composing approximately one-fourth or one-half of the patients, with 11% risk of failure and only 1% or 4% 5-year lymphoma-related mortality. Conclusion The combination of E/IV with either bulky disease or LDH ≥2 times upper limit of normal defined high-risk but not very-high-risk subgroups. More importantly, their absence defined subgroups comprising approximately one-fourth or one-half of the pts, with 11% risk of failure and minimal lymphoma-related mortality, who may not need more intensive treatment such as R-da-EPOCH. Implications for Practice By analyzing the impact of baseline clinical characteristics on outcomes of a large cohort of patients with primary mediastinal large B-cell lymphoma homogeneously treated with R-CHOP with or without radiotherapy, we developed novel prognostic indices which can aid in deciding which patients can be adequately treated with R-CHOP and do not need more intensive regimens such as R-da-EPOCH. The new indices consist of objectively determined characteristics (extranodal disease or stage IV, bulky disease, and markedly elevated serum lactate dehydrogenase), which are readily available from standard initial staging procedures and offer better discrimination compared with established risk scores (International Prognostic Index [IPI] and age-adjusted IPI).
- Published
- 2020
24. Over-accessible chromatin links myeloma initiating genetic events to oncogenic transcriptomes and aberrant transcription factor regulatory networks
- Author
-
Nikolaos Trasanidis, Evdoxia Hatjiharissi, Holger W. Auner, Xiaolin Xiao, Sudbery Im Im, Alexia Katsarou, Papaioannou M M, Irene Roberts, Maria Atta, Kanagaraju Ponnusamy, Aristeidis Chaidos, Jaime Alvarez-Benayas, Anastasios Karadimitris, Valentina S. Caputo, Marco Bua, and Philippa C. May
- Subjects
Transcriptome ,Genetic heterogeneity ,medicine ,Ectopic expression ,Computational biology ,Biology ,Enhancer ,medicine.disease ,Gene ,Transcription factor ,Multiple myeloma ,Chromatin - Abstract
Multiple myeloma is a genetically heterogeneous cancer of the bone marrow plasma cells (PC). Myeloma initiating genetic events define subgroups (MIE) and drive distinct oncogenic transcriptomes that converge into a mutually exclusive overexpression of CCND1 and CCND2 oncogenes. Here, with reference to normal PC, we dissect how MIE impact the chromatin regulatory landscape of MM. We find that chromatin accessibility combined with transcriptome profiling classifies myeloma genetic subgroups, while in a topologically constrained manner, distal rather than proximal regulatory elements influence myeloma transcriptomes. Across and within MIE-defined subgroups, genes and pathways critical for myeloma biology can be linked to developmentally activated or de novo formed enhancers. We show that existing transcription factors, co-opted to organise highly ordered, aberrant regulatory networks, generate known and novel myeloma cell dependencies and help identify prognostic markers. Finally, we discover and functionally validate the critical enhancer that regulates ectopic expression of CCND2 in MM.
- Published
- 2020
- Full Text
- View/download PDF
25. Safety and Efficacy of Daratumumab with Ixazomib and Dexamethasone in Patients with One Prior Lenalidomide-Based Regimen: Outcomes of the Phase 2 Daria Study
- Author
-
Argiris Symeonidis, Maria Gavriatopoulou, Magdalini Migkou, Sosana Delimpasi, Meletios A. Dimopoulos, Eirini Katodritou, Evdoxia Hatjiharissi, Alexandros Leonidakis, Efstathios Kastritis, Ioanna Dialoupi, Evgenia Verrou, and Evangelos Terpos
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Daratumumab ,Cell Biology ,Hematology ,Biochemistry ,Ixazomib ,Regimen ,chemistry.chemical_compound ,chemistry ,Internal medicine ,medicine ,In patient ,business ,health care economics and organizations ,Dexamethasone ,Lenalidomide ,medicine.drug - Abstract
Introduction The use of lenalidomide in frontline therapy for patients (pts) with newly diagnosed multiple myeloma (NDMM) has increased the number of pts who become refractory to lenalidomide at second line. Thus, there is an ongoing need for new treatment options for lenalidomide refractory patients at first relapse. Daratumumab, an anti-CD38 monoclonal antibody, has shown significant activity and acceptable safety in pts with relapsed/refractory multiple myeloma (RRMM) as monotherapy or in combination with other agents. Ixazomib, the first oral proteasome inhibitor, in combination with lenalidomide and dexamethasone also improves key survival endpoints in RRMM. This study assessed the efficacy of daratumumab in combination with ixazomib and dexamethasone (Dara-Ixa-dex) as second-line therapy in pts with RRMM who have been previously treated with a lenalidomide-based regimen. Methods DARIA is an ongoing, prospective, open-label, multicenter, phase 2 study. Eligible adult pts with RRMM had measurable disease after one prior line with a lenalidomide-based regimen and a Karnofsky Performance Status (KPS) score of ≥70. Exclusion criteria included previous anti-CD38 or ixazomib treatment, treatment with CYP3A inducers or use of St. John's wort within 14 days before C1D1, anti-myeloma treatment within 2 weeks or 5 pharmacokinetic half-lives of the treatment prior to C1D1 and allogenic or autologous stem cell transplantation (ASCT) within 12 weeks before C1D1. Treatment with Dara-Ixa-dex comprises an induction phase of nine 28-days cycles and a maintenance phase. In induction, pts received DARA 16mg/kg (weekly for cycles 1-2, bi-weekly for cycles 3-6, and every 4 weeks thereafter) administered intravenously until November 2020 and subcutaneously at a fixed dose of 1800 mg thereafter; 4mg oral ixazomib (days 1, 8, and 15 of each cycle); and 40mg oral dexamethasone (weekly, each cycle). In maintenance, Dara-Ixa were administered every 4 weeks until disease progression or unacceptable toxicity, with dexamethasone being discontinued. The primary endpoint was overall response rate (ORR). Secondary endpoints included progression-free survival (PFS), the toxicity profile of Dara-Ixa-dex, and the effects of the combination on serum bone metabolism markers (C-terminal telopeptide of type 1 collagen [CTX], tartrate-resistant acid phosphatase isoform 5b [TRACP-5b], bone-specific alkaline phosphatase [bALP], and osteocalcin [OC]) from baseline until disease progression. Results The study has completed accrual and 50 pts have been enrolled (mean [range] age: 69.0 (50.0-89.0) years; female: 28 [56.0%]). At screening, 24 (48.0%) pts had a KPS score ≥90, and most pts were at stage ≤II by the International Staging System (ISS; 42, 84.0%) or by the revised ISS (47, 94.0%). Thirty-two (64.0%) pts were refractory to lenalidomide, and 15 (37.5%) had prior ASCT. The median (range) number of study treatment cycles was 9 (1-26). ORR was 50.0% (25 pts); one (2.0%) pt had a complete response, 13 (26.0%) a very good partial response (VGPR), and 11 (22.0%) pts had a PR. The median (range) time from first Dara-Ixa-dex dose until first response (≥PR) is 1.0 (0.9-3.7) month. The median PFS was 8.1 months (95% CI: 5.8-15.6) (Figure). After a median (range) follow-up of 12.4 (0.9-28.4) months, 14 (28.0%) pts are still on treatment; reasons for treatment discontinuation were progressive disease (28 pts, 77.8%), physician's decision and fatal serious adverse event [SAE] (3 pts, 8.3% each), and adverse event [AE] (2pts, 5.6%). Following 15 months of treatment, the median change from baseline for CTX, TRACP-5b, bALP and OC were significant (p Overall, 20 (40.0%) pts have ≥1 grade 3/4 AE, the most common being thrombocytopenia (10 pts, 20.0%), and 14 (28.0%) pts have ≥1 SAE, the most common being acute kidney injury and pneumonia (2 pts [4.0%] each condition). Four fatal SAEs were reported (pneumonia, infection, urinary tract infection, and lower respiratory tract infection). Conclusions Second-line treatment with Dara-Ixa-dex in pts with RRMM who were pre-treated with a lenalidomide-based regimen resulted in rapid ( Figure 1 Figure 1. Disclosures Terpos: Genesis: Consultancy, Honoraria, Research Funding; BMS: Honoraria; Amgen: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; GSK: Honoraria, Research Funding; Janssen-Cilag: Consultancy, Honoraria, Research Funding; Novartis: Honoraria; Takeda: Consultancy, Honoraria, Research Funding; Sanofi: Consultancy, Honoraria, Research Funding. Gavriatopoulou: Sanofi: Honoraria; Genesis: Honoraria; GSK: Honoraria; Karyopharm: Honoraria; Amgen: Honoraria; Janssen: Honoraria; Takeda: Honoraria. Katodritou: GSK, Amgen, Karyopharm, Abbvie, Janssen-Cilag, Genesis Pharma, Sanofi: Honoraria, Research Funding. Hatjiharissi: Gilead: Honoraria; Genesis: Honoraria, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Verrou: Karyopharm: Research Funding; Takeda: Honoraria; Genesis: Honoraria; Abbvie: Honoraria, Research Funding; Janssen Cilag: Honoraria, Research Funding; Roche: Honoraria; Amgen: Honoraria. Leonidakis: Health Data Specialists: Current Employment. Delimpasi: Amgen: Honoraria, Speakers Bureau; Janssen: Honoraria, Speakers Bureau; Takeda: Honoraria, Speakers Bureau. Symeonidis: MSD: Consultancy, Research Funding; GSK: Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astellas: Consultancy, Research Funding; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GenesisPharma: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi/Genzyme: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Demo: Research Funding; WinMedica: Research Funding. Dimopoulos: Amgen: Honoraria; Janssen: Honoraria; BMS: Honoraria; Beigene: Honoraria; Takeda: Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
26. Efficacy and Safety of Daratumumab with Dexamethasone in Patients with Relapsed/Refractory Multiple Myeloma and Severe Renal Impairment or on Dialysis: Final Analysis of the Phase 2 Dare Study
- Author
-
Evangelos Terpos, Magdalini Migkou, Argiris Symeonidis, Michele Cavo, Efstathios Kastritis, Eirini Katodritou, Nikolaos Kanellias, Maria Gavriatopoulou, Sosana Delimpasi, Marie-Christine Kyrtsonis, Meletios A. Dimopoulos, Evdoxia Hatjiharissi, Alexandros Leonidakis, Maria Roussou, Despina Fotiou, Elena Rivolti, Elena Zamagni, and Kyriaki Manousou
- Subjects
medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,Urology ,Daratumumab ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Relapsed refractory ,medicine ,In patient ,business ,Multiple myeloma ,Dialysis ,Dexamethasone ,medicine.drug - Abstract
Introduction Despite the availability of novel agents in treating multiple myeloma (MM), renal impairment (RI) remains a poor prognostic factor, and the median survival of patients (pts) with MM and RI is approximately half of that for MM pts with normal renal function. RI can affect up to 50% of pts with MM at presentation, highlighting the need for effective treatment options for this patient population. Daratumumab, an IgG1 κ human monoclonal antibody that targets CD38, has shown efficacy and a favorable safety profile in pts with relapsed or refractory MM (RRMM). The DARE study assessed the safety and efficacy of daratumumab with dexamethasone in pts with RRMM and severe RI or requiring hemodialysis. Methods DARE is a prospective, open-label, phase 2 study, conducted in eight sites in Greece and Italy. Eligible pts were adults with documented RRMM and severe RI (estimated glomerular filtration rate [eGFR] Results The study has completed accrual and 38 pts were enrolled. The pts' median (range) age was 72 (40-89) years, and most pts were male (29, 76.3%). At baseline, 37 (97.4%) pts had ECOG PS ≤1 and 34 (89.5%) were at International Staging System (ISS) stage III. By revised ISS, 20 (52.6%) and 16 (42.1%) pts were at stages II and III, respectively. Pts had a median (range) of 3 (2-6) prior systemic therapies; thirteen (34.2%) pts had undergone prior autologous stem cell transplantation. The median eGFR at baseline was 13.0 mL/min/1.73m 2 and seventeen (44.7%) pts were on dialysis at the time of enrollment. The median (range) number of cycles given was 8.0 (1.0-38.0), and the median (range) follow-up was 11.3 ( Overall, 19 (50.0%) pts had ≥1 grade 3/4 adverse event (AE), and 10 (26.3%) had ≥1 serious AE (SAE). The most common grade 3/4 AEs were anemia (6 pts, 15.8%), hyperglycemia (5 pts, 13.2%), and hypercalcemia (3 pts, 7.9%). The most common SAE was septic shock (3 pts, 7.9%). Conclusions The administration of daratumumab with dexamethasone in pts with RRMM and severe RI or requiring hemodialysis is safe and effective therapy associated with a median PFS of approximately 12 months; hematologic responses were rapid and observed within one month from treatment initiation and approximately one-fifth of pts achieved a major renal response. Almost half of the pts were on dialysis and the combination was active and safe also for those on dialysis. No new safety signals were observed with daratumumab in pts with RRMM and severe RI or in need of dialysis. Figure 1 Figure 1. Disclosures Kastritis: Janssen: Consultancy, Honoraria, Research Funding; Takeda: Honoraria; Pfizer: Consultancy, Honoraria, Research Funding; Genesis Pharma: Honoraria; Amgen: Consultancy, Honoraria, Research Funding. Terpos: Sanofi: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; Novartis: Honoraria; Janssen-Cilag: Consultancy, Honoraria, Research Funding; GSK: Honoraria, Research Funding; Genesis: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; BMS: Honoraria; Amgen: Consultancy, Honoraria, Research Funding. Symeonidis: Demo: Research Funding; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; WinMedica: Research Funding; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; GenesisPharma: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GSK: Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; MSD: Consultancy, Research Funding; Sanofi/Genzyme: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astellas: Consultancy, Research Funding; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Delimpasi: Janssen: Honoraria, Speakers Bureau; Amgen: Honoraria, Speakers Bureau; Takeda: Honoraria, Speakers Bureau. Cavo: Bristol-Myers Squib: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: TRAVEL, ACCOMMODATIONS, EXPENSES, Speakers Bureau; Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; GlaxoSmithKline: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Adaptive Biotechnologies: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel Accommodations, Speakers Bureau; Novartis: Honoraria. Zamagni: Janssen: Honoraria; Bristol-Myers-Squibb: Honoraria; Takeda: Honoraria; Amgen: Honoraria. Katodritou: GSK, Amgen, Karyopharm, Abbvie, Janssen-Cilag, Genesis Pharma, Sanofi: Honoraria, Research Funding. Kyrtsonis: Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene/Genesis Pharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Honoraria; Sanofi: Membership on an entity's Board of Directors or advisory committees. Hatjiharissi: Gilead: Honoraria; Genesis: Honoraria, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Leonidakis: Health Data Specialists: Current Employment. Manousou: Health Data Specialists: Current Employment. Gavriatopoulou: Sanofi: Honoraria; Genesis: Honoraria; Amgen: Honoraria; Janssen: Honoraria; GSK: Honoraria; Karyopharm: Honoraria; Takeda: Honoraria. Dimopoulos: Amgen: Honoraria; Beigene: Honoraria; Janssen: Honoraria; BMS: Honoraria; Takeda: Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
27. Impact of Daratumumab Monotherapy on Bone Metabolism Parameters in Patients with Relapsed and/or Refractory Multiple Myeloma Who Have Received at Least Two Prior Lines of Therapy Including Lenalidomide and a Proteasome Inhibitor: Outcomes of the Phase 2 Rebuild Study
- Author
-
Marie-Christine Kyrtsonis, Maria Gavriatopoulou, Sosana Delimpasi, Evgenia Verrou, Evangelos Eleutherakis Papaiakovou, Argiris Symeonidis, Evangelos Terpos, Maria Papaioannou, Efstathios Kastritis, Eirini Katodritou, Meletios A. Dimopoulos, Evdoxia Hatjiharissi, Alexandros Leonidakis, and Ioannis Ntanasis-Stathopoulos
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Daratumumab ,Refractory Multiple Myeloma ,Cell Biology ,Hematology ,Biochemistry ,Bone remodeling ,Internal medicine ,medicine ,Proteasome inhibitor ,In patient ,business ,health care economics and organizations ,Lenalidomide ,medicine.drug - Abstract
Introduction: Osteolytic bone disease is the most common clinical feature of multiple myeloma (MM), affecting approximately 80% of patients (pts) at diagnosis. Non-invasive biomarkers of bone resorption and formation are indicative of bone dynamics and have been used to assess bone metabolism during anti-myeloma treatment. Daratumumab (dara), an IgG1 κ human monoclonal antibody targeting CD38, is approved as treatment of patients with relapsed and/or refractory MM (RRMM). Moreover, dara inhibits in vitro osteoclastogenesis and bone resorption. The present study investigated the impact of dara monotherapy on bone remodeling in pts with RRMM who have had ≥2 prior lines of therapy. Methods: REBUILD was a prospective, non-comparative, open-label, phase 2 study, conducted in six centers in Greece. Eligible pts were adults with documented RRMM, who have had ≥2 prior lines of therapy (including lenalidomide and a PI), evidence of progressive disease by International Myeloma Working Group criteria, a Karnofsky Performance Status score ≥70, and creatinine clearance ≥30 mL/min. Exclusion criteria included previous treatment with an anti-CD38 antibody, such as dara. Pts received intravenous dara at a 16 mg/kg dose weekly for 8 weeks, then every 2 weeks for an additional 16 weeks, followed by 4 weeks interval thereafter. The primary endpoint was the change from baseline in the bone resorption markers C-terminal cross-linking telopeptide of type 1 collagen (CTX) and tartrate-resistant acid phosphatase isoform 5b (TRACP-5b) after 4 months of dara monotherapy. Secondary endpoints included the change from baseline to 4 months of treatment in selected bone formation markers (e.g., bone-specific alkaline phosphatase [bALP], osteocalcin [OC], and procollagen type-I N-propeptide [PINP]), markers of osteoclast regulation (RANKL, osteoprotegerin, dicckopf-1 [DKK-1], sclerostin and C-C motif ligand-3 [CCL3]), progression-free survival (PFS), and overall survival (OS). Results: Among the 57 pts enrolled in the study, 33 (57.9%) pts had biomarker and other clinical data available on treatment initiation (baseline) and after 4 months of treatment; the following analyses refer to this pt subgroup. The pts' median (range) age was 73.0 (52.0-84.0) years, and most were female (18, 54.5%). Most (16, 48.5%) pts had >10 lytic bone lesions at baseline. Six (18.2%) pts received bisphosphonates along with dara monotherapy. The ORR (Partial Response or better [≥PR]) was 63.6% (CR: 3.0%, VGPR: 21.2%, PR: 39.4%). The respective 4-month ORR was 57.6% (VGPR: 24.2%, PR: 33.3%) The median CTX change from baseline to 4 months was 3.9%, with 13 (39.4%) and 11 (33.3%) pts exhibiting ≥20% and ≥30% reduction in CTX levels, respectively. The TRACP-5b levels decreased from baseline to 4 months by a median of 2.6%, with 10 (30.3%) and 6 (18.2%) pts showing ≥20% and ≥30% reduction in TRACP-5b levels, respectively. The median changes from baseline to 4 months in the CTX and TRACP-5b levels for pts with a ≥PR at 4 months were -1.3% and -2.6%, respectively; the respective changes for pts not achieving favorable response (i.e., pts with minimal response, stable disease, disease progression, or no response assessment) were 5.3% and -7.2%. The levels of the bone metabolism biomarkers bALP, OC, and PINP increased from baseline to 4 months, the median changes being 18.4%, 92.6% and 10.2%, respectively. For pts with ≥PR at 4 months, the median changes from baseline to 4 months in bALP, OC, and PINP levels were 25.3%, 146.0% and 15.7%, respectively; the respective changes for pts not achieving favourable response were 18.3%, 15.6% and -7.3% (Table 1). Other significant differences at 4 months included the decrease in DKK-1 by a median of 17.5%, the decrease in CCL3 by 16.0% (Table 1). The median (95% confidence interval) PFS and OS were 9.3 (6.7-15.3) and 21.2 (11.4-not reached) months, respectively; the respective results for all 57 patients were 4.7 (3.0-7.2) and 10.5 (8.4-18.1) months. Conclusions: In these highly pre-treated MM pt group, dara monotherapy showed a positive effect on bone metabolism which was more profound among responders; OC had a ~3-fold increase after 4 months of therapy. The reduction in TRACP-5b and in CCL-3 but not in CTX suggests a mild inhibitory effect on osteoclasts by dara, with a statistically non-significant trend to be greater in responders. Figure 1 Figure 1. Disclosures Terpos: Novartis: Honoraria; Takeda: Consultancy, Honoraria, Research Funding; Sanofi: Consultancy, Honoraria, Research Funding; Genesis: Consultancy, Honoraria, Research Funding; GSK: Honoraria, Research Funding; Janssen-Cilag: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; BMS: Honoraria; Amgen: Consultancy, Honoraria, Research Funding. Kastritis: Janssen: Consultancy, Honoraria, Research Funding; Takeda: Honoraria; Pfizer: Consultancy, Honoraria, Research Funding; Genesis Pharma: Honoraria; Amgen: Consultancy, Honoraria, Research Funding. Hatjiharissi: Gilead: Honoraria; Genesis: Honoraria, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Katodritou: GSK, Amgen, Karyopharm, Abbvie, Janssen-Cilag, Genesis Pharma, Sanofi: Honoraria, Research Funding. Verrou: Amgen: Honoraria; Genesis: Honoraria; Abbvie: Honoraria, Research Funding; Takeda: Honoraria; Roche: Honoraria; Janssen Cilag: Honoraria, Research Funding; Karyopharm: Research Funding. Gavriatopoulou: GSK: Honoraria; Karyopharm: Honoraria; Takeda: Honoraria; Janssen: Honoraria; Amgen: Honoraria; Genesis: Honoraria; Sanofi: Honoraria. Leonidakis: Health Data Specialists: Current Employment. Delimpasi: Janssen: Honoraria, Speakers Bureau; Takeda: Honoraria, Speakers Bureau; Amgen: Honoraria, Speakers Bureau. Kyrtsonis: Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene/Genesis Pharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Honoraria; Sanofi: Membership on an entity's Board of Directors or advisory committees. Symeonidis: AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi/Genzyme: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GSK: Research Funding; MSD: Consultancy, Research Funding; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Demo: Research Funding; Astellas: Consultancy, Research Funding; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; WinMedica: Research Funding; GenesisPharma: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Dimopoulos: Beigene: Honoraria; Janssen: Honoraria; BMS: Honoraria; Amgen: Honoraria; Takeda: Honoraria.
- Published
- 2021
- Full Text
- View/download PDF
28. Efficacy and Tolerability of Daratumumab with Ixazomib and Dexamethasone in Patients with One Prior Lenalidomide-Based Regimen: Preliminary Results of the Phase 2 Daria Study
- Author
-
Ioanna Dialoupi, Maria Gavriatopoulou, Efstathios Kastritis, Evgenia Verrou, Sosana Delimpasi, Evangelos Terpos, Argiris Symeonidis, Evdoxia Hatjiharissi, Alexandros Leonidakis, Magdalini Migkou, Meletios A. Dimopoulos, and Eirini Katodritou
- Subjects
medicine.medical_specialty ,business.industry ,education ,Immunology ,Bone markers ,Daratumumab ,Cell Biology ,Hematology ,Biochemistry ,Ixazomib ,Health data ,Regimen ,chemistry.chemical_compound ,Tolerability ,chemistry ,Family medicine ,medicine ,In patient ,business ,health care economics and organizations ,Lenalidomide ,medicine.drug - Abstract
Introduction: Lenalidomide-based regimens are currently among the standard-of-care treatment options for newly diagnosed patients with multiple myeloma NDMM). Second-line treatment of patients who are refractory to lenalidomide or have relapsed after prior use of lenalidomide is challenging. Daratumumab, an IgG1κ human monoclonal antibody that targets CD38, has shown durable responses and a favorable safety profile in heavily pretreated patients with relapsed/refractory MM (RRMM) as monotherapy, or in combination with other anti-myeloma treatments. Ixazomib, the first oral proteasome inhibitor, has shown deep responses and a positive safety profile in NDMM patients when it is combined with lenalidomide and dexamethasone. The aim of the DARIA study is to evaluate the effectiveness of daratumumab in combination with ixazomib and dexamethasone (DId) as second-line therapy in patients who have received prior treatment with lenalidomide-based regimens. Methods: DARIA is an ongoing prospective, open-label, multicenter, phase 2 study, which aims to enroll 43 patients who have received one prior line of therapy with a lenalidomide-based regimen. Patients must have a Karnofsky performance status (PS) ≥70. Exclusion criteria include previous daratumumab or anti-CD38, or ixazomib treatment exposure. The treatment phase consists of an induction therapy for 9 cycles, followed by a maintenance period. The induction phase included 28-day treatment cycles with 16 mg/kg intravenous daratumumab (weekly for cycles 1-2, every 2 weeks for cycles 3-6, and every 4 weeks thereafter), 4 mg oral ixazomib (Days 1, 8, and 15 of each cycle), and 40 mg oral dexamethasone for the first 9 cycles (weekly, each cycle). During the maintenance phase of the study daratumumab is administered every 4 weeks and ixazomib on the same schedule, until disease progression or unacceptable toxicity whereas dexamethasone is discontinued. The primary endpoint is the overall response rate (ORR). Secondary endpoints include evaluation of the toxicity profile of the study treatment, progression-free survival (PFS), overall survival, and levels of serum bone markers and angiogenic cytokines. All responses are based on investigators' assessment per International Myeloma Working Group criteria. This analysis presents results for patients who received the first dose of study treatment ≥5 months prior to the cut-off date (01/05/2020). Results: Fifteen patients, enrolled in 4 Sites, are included in the current analysis. Overall, 60% of the patients were refractory to lenalidomide. Median age was 70 years and the majority of patients were male (60%). The median time from diagnosis to first dose of study treatment was 1.3 years. Two patients (13%) had a prior autologous stem cell transplantation. Patients had a Karnofsky PS of 70 (27%), 80 (7%), 90 (53%), and 100 (13%) at baseline. At screening 47%, 26%, and 27% of the patients had an international staging system (ISS) of 1, 2, and 3, respectively, and a revised ISS of 1 (21%), 2 (71%), and 3 (7%). The median number of cycles reached until the cut-off date was 7. The median time from first dose of study treatment until first partial (PR) or very good partial response (VGPR) was 0.9 month. ORR was 60%, with 47% of the patients achieving VGPR and 13% PR. The 12-month PFS rate was 54%, and 8 patients (53%) were still on treatment by the cut-off date. From the patients who discontinued 71% was due to disease progression and 29% due to physician's decision. Overall, 10 patients (67%) had at least one adverse event (AE) grade 3 or 4, the most common being thrombocytopenia (6 patients, 40%). Four patients (27%) had a single SAE each: lower respiratory tract infection (fatal); peritonitis, gastroenteritis; nephrolithiasis. Conclusions: Rapid (median time to PR or better was 1 month) and deep responses (47% of the patients exhibited VGPR) were observed following treatment with daratumumab, ixazomib and dexamethasone as second-line therapy in patients who were previously treated with a lenalidomide-based regimen, more than half of whom were refractory to lenalidomide. The safety profile of DId combination is very good with one third of the patients not experiencing any grade 3 or 4 AEs, when at the same time the PFS rate at 12 months reached 54%. Disclosures Terpos: Janssen: Honoraria, Research Funding; Takeda: Honoraria, Other: travel expenses , Research Funding; Celgene: Honoraria; Amgen: Honoraria, Research Funding; Genesis pharma SA: Honoraria, Other: travel expenses , Research Funding; Sanofi: Honoraria; BMS: Honoraria. Gavriatopoulou:Amgen: Consultancy, Honoraria; Karyopharm: Consultancy, Honoraria; Genesis Pharma: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria. Katodritou:Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Karyopharm: Research Funding; Abbvie: Research Funding; Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Theagenion Cancer Hospital: Current Employment; Takeda: Honoraria, Other: Expenses, Research Funding; Genesis Pharma: Honoraria, Other: Expenses, Research Funding. Hatjiharissi:Abbvie: Honoraria; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria. Verrou:Amgen: Honoraria; Abbvie: Honoraria; Janssen Cilag: Honoraria, Research Funding; Karyopharm: Research Funding; Takeda: Honoraria; Roche: Honoraria; Genesis: Honoraria. Leonidakis:Health Data Specialists S.A.: Current Employment. Delimpasi:Janssen: Honoraria; Takeda: Honoraria; Amgen: Honoraria; GENESIS: Honoraria. Symeonidis:Sanofi/Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GenesisPharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Research Funding; WinMedica: Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck Sharp & Dohme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astellas: Research Funding. Kastritis:Amgen: Consultancy, Honoraria, Research Funding; Pfizer: Consultancy, Honoraria; Genesis Pharma: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria. Dimopoulos:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Speakers Bureau; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees.
- Published
- 2020
- Full Text
- View/download PDF
29. Daratumumab with Dexamethasone in Patients with Relapsed/Refractory Multiple Myeloma and Severe Renal Impairment: Results on Efficacy and Safety of the Phase 2 Dare Study
- Author
-
Efstathios Kastritis, Maria Roussou, Nikolaos Kanellias, Magdalini Migkou, Marie-Christine Kyrtsonis, Despina Fotiou, Eirini Katodritou, Evangelos Terpos, Meletios A. Dimopoulos, Sosana Delimpasi, Elena Rivolti, Elena Zamagni, Argiris Symeonidis, Maria Gavriatopoulou, Michele Cavo, and Evdoxia Hatjiharissi
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Daratumumab ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Internal medicine ,Relapsed refractory ,medicine ,In patient ,business ,Multiple myeloma ,Dexamethasone ,medicine.drug - Abstract
Introduction: Renal impairment (RI) is common in multiple myeloma (MM), with up to 40% of the patients (pts) experiencing this complication during the course of their disease. RI increases risk of early death and affects disease management in multiple ways, as it may complicate treatment options and dosing, and render pts more susceptible to infections and prolonged hospitalizations. Therefore, there is an urgent need to restore renal function in these pts in order to improve their quality of life and prognosis. Bortezomib-based therapies are the most commonly used in pts with RI, but eventually pts may become refractory to bortezomib and other drug classes such as IMIDs. Thus, new therapeutic options are needed in order to manage pts with MM and RI who fail these drugs. Daratumumab, an IgG1κ human monoclonal antibody that targets CD38, has shown durable responses and a favorable safety profile in heavily pretreated pts with relapsed/refractory MM (RRMM) as monotherapy. Pharmacokinetic analyses suggest that there are no clinically important differences in exposure to daratumumab in pts with normal or impaired renal function, but the available data on safety and efficacy of pts with RRMM and severe RI is scarce. Methods: DARE is a prospective, open-label, multicenter, phase 2 study, which completed the enrolment of 38 adult pts with documented RRMM and severe RI, defined as either eGFR Results: The current analysis includes 35 pts, enrolled in 7 Sites. Median age was 72 years, and 77.1% were male. The median time from diagnosis to first daratumumab dose was 4.2 years. Pts had a median of 3 prior systemic therapies, and 37.1% had a prior autologous stem cell transplantation. At study initiation 8.6% and 91.4% of pts had international staging system (ISS) 2 and 3 disease, respectively, while 51.4% and 48.6% were revised ISS 2 and 3. The median eGFR at baseline was 13 mL/min/1.73 m2.and 17 pts (48.6%) were on dialysis. The median number of cycles administered until the cut-off date was 5 and the median follow-up duration was 5.5 months. The 6-month progression-free survival rate, was 50% (figure). Overall, ORR was 45.7%, with 31.4% of all pts achieving a VGPR and 14.3% a PR. For pts on dialysis (n=17), ORR was 35.3%, equally divided between pts achieving VGPR and PR (17.6%). The median time from the first dose of study treatment until the first response (≥PR) was 0.9 months. RRR was 17.1%. By the cut-off date, 37.1% of the pts were still receiving protocol therapy, 17.1% discontinued treatment due to death, and 31.4% due to disease progression. Overall, 17 pts (48.6%) had at least 1 adverse event (AE) of grade 3 or 4, most frequent being anemia (17.1%) and hyperglycemia (8.6%). Nine (25.7%) pts had at least 1 SAE: Septic shock (fatal, 2 pts), and performance status decreased (fatal), lower respiratory tract infection (fatal), myocardial infarction (fatal), peritonitis (fatal, in a patient receiving peritoneal dialysis), cerebrovascular accident, pneumonia, acute kidney injury, and hyperkalemia (1 pt each). Conclusions: The administration of daratumumab with dexamethasone led to rapid hematologic responses in pts with RRMM and severe RI, including those in dialysis, and at the same time it resulted in a major renal response for 17.1% of the pts. Importantly, new safety signals were not observed, and daratumumab can be safely administered in pts with severe RI or those on dialysis. Figure 1 Disclosures Kastritis: Janssen: Consultancy, Honoraria, Research Funding; Genesis Pharma: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Research Funding. Terpos:Celgene: Honoraria; Takeda: Honoraria, Other: travel expenses , Research Funding; Janssen: Honoraria, Research Funding; Genesis pharma SA: Honoraria, Other: travel expenses , Research Funding; Amgen: Honoraria, Research Funding; Sanofi: Honoraria; BMS: Honoraria. Symeonidis:Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Research Funding; WinMedica: Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astellas: Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GenesisPharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck Sharp & Dohme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi/Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Delimpasi:Genesis: Honoraria; Takeda: Honoraria; Janssen: Honoraria; Amgen: Honoraria. Cavo:Jannsen, BMS, Celgene, Sanofi, GlaxoSmithKline, Takeda, Amgen, Oncopeptides, AbbVie, Karyopharm, Adaptive: Consultancy, Honoraria. Zamagni:BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses, Speakers Bureau; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda: Honoraria, Other: Travel, Accommodations, Expenses, Speakers Bureau; Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses, Speakers Bureau; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Travel, Accommodations, Expenses, Speakers Bureau; Celgene Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Katodritou:Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genesis Pharma: Honoraria, Other: Expenses, Research Funding; Abbvie: Research Funding; Karyopharm: Research Funding; Theagenion Cancer Hospital: Current Employment; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Honoraria, Other: Expenses, Research Funding. Rivolti:Celgene: Membership on an entity's Board of Directors or advisory committees. Kyrtsonis:Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees. Gavriatopoulou:Amgen: Consultancy, Honoraria; Karyopharm: Consultancy, Honoraria; Genesis Pharma: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria. Hatjiharissi:Abbvie: Honoraria; Gilead: Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria; Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Dimopoulos:Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Speakers Bureau; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees.
- Published
- 2020
- Full Text
- View/download PDF
30. The Stat3/5 Signaling Biosignature in Hematopoietic Stem/Progenitor Cells Predicts Response and Outcome in Myelodysplastic Syndrome Patients Treated with Azacitidine
- Author
-
Costas Tsatalas, Athanasios Galanopoulos, Eleftheria Lamprianidou, Vasiliki Pappa, Evangelia Nakou, Maria Papaioannou, Theodoros P. Vassilakopoulos, Evdoxia Hatjiharissi, Andreas Scorilas, Christos K. Kontos, Sotirios G. Papageorgiou, Vassilia Garypidou, Ioannis Kotsianidis, Paraskevi Miltiades, Panagiotis G. Adamopoulos, Helen A. Papadaki, Sofia Vakalopoulou, and Emmanouil Spanoudakis
- Subjects
Male ,STAT3 Transcription Factor ,0301 basic medicine ,Antimetabolites, Antineoplastic ,Cancer Research ,Myeloid ,Proteome ,Azacitidine ,Biology ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Granulocyte Colony-Stimulating Factor ,STAT5 Transcription Factor ,medicine ,Cluster Analysis ,Humans ,Progenitor cell ,Aged ,Aged, 80 and over ,Myelodysplastic syndromes ,Myeloid leukemia ,Middle Aged ,Hematopoietic Stem Cells ,Prognosis ,medicine.disease ,Survival Analysis ,Leukemia, Myeloid, Acute ,Leukemia ,Haematopoiesis ,Phenotype ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Immunology ,Neoplastic Stem Cells ,Cancer research ,Female ,Biomarkers ,Signal Transduction ,medicine.drug - Abstract
Purpose: Azacitidine is the mainstay of high-risk myelodysplastic syndromes (MDS) therapy, but molecular predictors of response and the mechanisms of resistance to azacitidine remain largely unidentified. Deregulation of signaling via Stat3 and Stat5 in acute myeloid leukemia (AML) is associated with aggressive disease. Numerous genes involved in cell signaling are aberrantly methylated in MDS, yet the alterations and the effect of azacitidine treatment on Stat3/5 signaling in high-risk MDS have not been explored. Experimental Design: We assessed longitudinally constitutive and ligand-induced phospho-Stat3/5 signaling responses by multiparametric flow cytometry in 74 patients with MDS and low blast count AML undergoing azacitidine therapy. Pretreatment Stat3/5 signaling profiles in CD34+ cells were grouped by unsupervised clustering. The differentiation stage and the molecular properties of the CD34+ G-CSF–inducible Stat3/5 double-positive subpopulation were performed by flow cytometry and quantitative real-time PCR in isolated MDS progenitors. Results: The pretreatment Stat3/5 signaling profiles in CD34+ cells correlated strongly with response and cytogenetics and independently predicted event-free survival. We further identified a CD34+ G-CSF–inducible Stat3/5 double-positive subpopulation (DP subset) whose pretreatment levels were inversely associated with treatment response and cytogenetics. The kinetics of the DP subset followed the response to azacitidine and the disease course, whereas its molecular characteristics and cellular hierarchy were consistent with a leukemia propagating cell phenotype. Conclusions: Our findings provide a novel link among Stat3/5 signaling and MDS pathobiology and suggest that the Stat3/5 signaling biosignature may serve as both a response biomarker and treatment target. Clin Cancer Res; 22(8); 1958–68. ©2015 AACR.
- Published
- 2016
- Full Text
- View/download PDF
31. T Cell Immunoprofiling of Patients with Relapsed and/or Refractory Myeloma Who Receive Daratumumab Monotherapy: Longitudinal Analysis during 7 Cycle Follow-up of the Rebuild Phase 2 Study
- Author
-
Alexandra Siorenta, Elisavet Vlachonikola, Konstantia Kotta, Efstathios Kastritis, Maria Gavriatopoulou, Meletios A. Dimopoulos, Kostas Stamatopoulos, Maria Karipidou, Eirini Katodritou, Anastasia Chatzidimitriou, Argiris Symeonidis, Evangelos Terpos, Marina Gerousi, Evdoxia Hatjiharissi, Electra Sofou, and Sosana Delimpasi
- Subjects
medicine.medical_specialty ,business.industry ,Immunology ,Daratumumab ,Context (language use) ,Cell Biology ,Hematology ,Biochemistry ,Peripheral blood ,Median frequency ,Partial response ,Family medicine ,medicine ,Current employment ,Post treatment ,business ,After treatment - Abstract
Preliminary evidence for T cell receptor (TR) repertoire renewal and increased TR clonality has been reported by our group (Vlachonikola et al., ASH 2019) in multiple myeloma (MM) patients (pts) receiving daratumumab monotherapy within the context of the REBUILD study, an ongoing prospective, multicenter, non-comparative, open-label, phase II study in pts with relapsed and/or refractory MM (RRMM) who have had ≥2 prior lines of therapy, including lenalidomide and a proteasome inhibitor. Herein, we report the results from the longitudinal analysis of the TR repertoire employing next generation sequencing (NGS) and multi-color flow cytometry in 24 pts who completed 3 cycles (n=24) and 6 cycles (n=11/24) of daratumumab monotherapy, in order to assess the immunomodulatory effects of daratumumab. We assessed 59 peripheral blood samples collected at screening (SCR, n=24), on Day 1 of Cycle 4 (C4, n=24) and Day 1 of Cycle 7 (C7, n=11). Patients were grouped based on best responses at C4 into responders (i.e. pts with partial response [PR, n=7] and very good PR [VGPR, n=8]), and non-responders (i.e. pts with minimal response [MR, n=2], stable disease [SD, n=5], or progressive disease [PD, n=2]). TRBV-TRBD-TRBJ gene rearrangements were subjected to paired-end NGS and raw reads (n=13,886,646 | median 239,969/sample) were processed through a purpose-built bioinformatics pipeline. Productive TRBV-TRBD-TRBJ rearrangements were taken into consideration (n=6,324,986 | median 100,738/sample) for the computation of clonotypes (i.e. TRB rearrangements with identical TRBV gene usage and amino acid complementarity-determining region 3 sequence). Overall, 325,789 distinct clonotypes (median 4,535 clonotypes/sample) were analyzed. The TR repertoire displayed clonal T cell expansions in both groups (responders/non-responders) in all pre/post-treatment timepoints. Clonality increased after treatment for both responders and non-responders in all assessed timepoints, with statistical significance at C4 in both groups (median cumulative frequency of the 10 most expanded T cell clonotypes/sample in responders: 31.6% pre-treatment vs 43% C4 post-treatment, p=0.009; and, in non-responders: 19.8% pre-treatment vs 39.6% C4 post-treatment, p=0.009). In both groups, the clonotype repertoire appeared to be renewed. Interestingly, in the responders' group a significant shift was noticed in the major clonotype repertoire at screening vs C4. In particular, the 10 most expanded clonotypes/sample at C4 represented expansions of clonotypes present at very low frequency at screening, whereas the most expanded clonotypes at screening decreased or even diminished post-treatment. Additionally, although the major post-treatment clonotype at C4 also dominated at C7 in most cases, certain lower frequency clonotypes at C4 emerged among the top-10 at C7. Thirteen shared clonotypes were identified amongst the post-treatment repertoires of different patients but not in other entities in public databases, raising the possibility that they may be "MM-specific" and selected by common MM-associated antigens. Finally, flow cytometry analysis revealed a significant increase post treatment in the percentage of CD3+ T cells (median frequency at SCR 63% vs 78.7% at C4 | p=0.0045 and 87.4% at C7 | p=0.0009), driven mostly by the expansion of the CD8+ T cell compartment (median frequency at SCR 31.4% vs 45.3% at C4 | p=0.0045 and 53.8% at C7 | p=0.001) in both groups. In conclusion, we document T cell clonal expansions and clonal drift after daratumumab treatment in MM. Our results suggest that daratumumab acts through renewing the greatest part of the pre-treatment TR clonotype repertoire, suggesting dynamic changes of the T cell compartment under treatment, a claim also supported by the significant increase in CD8+ cytotoxic T cell numbers overtime. The significant post-treatment expansion of certain low-frequency pre-treatment clones in responders raises the intriguing hypothesis that daratumumab treatment may have led to the outgrowth of anti-MM T cell clones, arguably contributing to clinical response. Disclosures Kastritis: Amgen: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Genesis Pharma: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria. Hatjiharissi:Abbvie: Honoraria; Gilead: Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Honoraria. Katodritou:Theagenion Cancer Hospital: Current Employment; Takeda: Honoraria, Other: Expenses, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Genesis Pharma: Honoraria, Other: Expenses, Research Funding; Abbvie: Research Funding; Karyopharm: Research Funding; Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Gavriatopoulou:Amgen: Consultancy, Honoraria; Karyopharm: Consultancy, Honoraria; Genesis Pharma: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria. Delimpasi:GENESIS: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Amgen: Consultancy, Honoraria. Symeonidis:Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi/Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Research Funding; Astellas: Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GenesisPharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck Sharp & Dohme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; WinMedica: Research Funding. Stamatopoulos:AstraZeneca: Honoraria; Janssen, Gilead, Abbvie: Honoraria, Research Funding. Dimopoulos:Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Speakers Bureau; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau. Terpos:Amgen: Honoraria, Research Funding; Genesis pharma SA: Honoraria, Other: travel expenses , Research Funding; Janssen: Honoraria, Research Funding; Takeda: Honoraria, Other: travel expenses , Research Funding; Celgene: Honoraria; Sanofi: Honoraria; BMS: Honoraria. Chatzidimitriou:Janssen: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
32. Efficacy of Daratumumab Monotherapy on Bone Metabolism of Patients with Advanced Relapsed/Refractory Multiple Myeloma: Results from the Phase 2 Rebuild Study
- Author
-
Maria Papaioannou, Evgenia Verrou, Evangelos Terpos, Eirini Katodritou, Ioannis Ntanasis-Stathopoulos, Evdoxia Hatjiharissi, Alexandros Leonidakis, Argiris Symeonidis, Maria Gavriatopoulou, Efstathios Kastritis, Sosana Delimpasi, Marie-Christine Kyrtsonis, Meletios A. Dimopoulos, and Evangelos Eleutherakis-Papaiakovou
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,education ,Immunology ,Daratumumab ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Bone remodeling ,Internal medicine ,Relapsed refractory ,medicine ,business ,health care economics and organizations ,Multiple myeloma - Abstract
Introduction: Osteolytic lesions (OL) is a devastating characteristic of multiple myeloma (MM) that decreases patients (pts) quality of life. The interaction between MM plasma cells and the bone microenvironment leads to intracellular and intercellular pathways that adversely alter the delicate balance between bone formation and bone resorption. Pre-clinical studies have shown that anti-CD38 therapy inhibit osteoclast formation. Daratumumab (dara), an IgG1κ human monoclonal antibody that targets CD38, has shown deep hematological responses in heavily pre-treated pts with relapsed/refractory MM (RRMM) as monotherapy. The aim of the REBUILD study is to evaluate the effect of dara monotherapy on bone metabolism in advanced RRMM pts. Methods: REBUILD is a prospective, open-label, multicenter, phase 2 study which has completed the enrolment of 57 pts with documented RRMM and ≥2 prior lines of therapy, including lenalidomide and a proteasome inhibitor. Pts had a Karnofsky Performance Status score of ≥70, and a creatinine clearance of ≥30 mL/min. Exclusion criteria included previous treatment with dara or other anti-CD38 therapy. Pts receive dara at a weekly dose of 16 mg/kg for Cycles 1-2, every 2 weeks for Cycles 3-6 and every 4 weeks thereafter. The primary endpoint of this study was the change from baseline in the bone resorption markers C-terminal telopeptide of collagen type I (CTX) and tartrate-resistant acid phosphatase isoform 5b (TRACP-5b) after 4 months of dara monotherapy. Secondary endpoints include the change at 4 months from baseline in bone formation markers (bone-specific alkaline phosphatase [bALP], osteocalcin [OC], and procollagen type-I N-propeptide [PINP]); markers of osteoclast regulation (receptor activator of nuclear factor kappa-B ligand [RANKL], osteoprotegerin [OPG] and chemokine (C- C motif) ligand-3 [CCL-3]); markers of osteoblast control (sclerostin [SCL], dickkopf-1 [DKK-1]), and progression-free survival (PFS). This preliminary analysis included pts who received the first dose of study treatment at least 5 months before the cut-off date (01/05/2020). Results: Fifty-one pts were enrolled in 6 sites; among them 29 pts had bone markers and clinical data available on baseline and after 4 months of study treatment and are included in the present analysis. Median age was 73 years, and approximately half of them were female (15 pts, 52%). Median number of previous therapies was 3 (range: 2-5). Fifteen pts (52%) had >10 osteolytic lesions at study initiation, and only 4 pts (14%) received bisphosphonates together with dara monotherapy. The median changes in CTX and TRACP-5b levels for all pts (n=29) after 4 months in study treatment were 3.9% and -2.2%, respectively. Overall, 10 pts (35%) had ≥ 30% reduction in CTX and 5 pts (17%) in TRACP-5b levels. The median changes after 4 months of dara monotherapy for pts with a response [pts who achieved partial response or better (≥PR; n=18, 62%)] were 3% for CTX and -3% for TRACP-5b; among pts with no response [minimal response, stable disease, disease progression, or no response assessment prior to death (n=11; 38%)] 4% for CTX, and 9% for TRACP-5b. The median changes in bone formation markers for all pts after 4 months of dara were 24.5% for bALP, 116.8% for OC, and 15.7% for PINP. The differences for pts with a response versus pts without a response were respectively 26% versus 18% for bALP, 190% versus -61% (p=0.020) for OC, and 22% versus -3% for PINP. Other major differences in 4 months of dara monotherapy were the decrease in DKK-1 by 49% in pts with a response versus 2% increase in pts with no response, and the decrease in CCL3 by 15% in pts with response versus 101% increase in pts with no response (p=0.039). The median PFS for all 51 pts was 4.6 months (95% CI: 2.8-7.2). Conclusions: Monotherapy with dara has a positive effect on bone metabolism even in these highly pre-treated pts with MM. Reduction of TRACP-5b and of CCL-3 in responsive pts suggests an inhibitory effect on osteoclasts by dara. Interestingly, we found that there is a strong bone formation effect in all pts treated with dara monotherapy, especially in those who responded to therapy, i.e. OC had a 2-fold increase after 4 months of therapy. This is at least partially due to the reduction of DKK-1 (osteoblast inhibitor) in responding patients. Based on these positive results a preclinical study on the effect of dara on bone cells is currently being performed. Disclosures Terpos: Amgen: Honoraria, Research Funding; BMS: Honoraria; Sanofi: Honoraria; Celgene: Honoraria; Takeda: Honoraria, Other: travel expenses , Research Funding; Janssen: Honoraria, Research Funding; Genesis pharma SA: Honoraria, Other: travel expenses , Research Funding. Kastritis:Janssen: Consultancy, Honoraria, Research Funding; Genesis Pharma: Consultancy, Honoraria; Takeda: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria; Amgen: Consultancy, Honoraria, Research Funding. Hatjiharissi:Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Roche: Honoraria; Abbvie: Honoraria; Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Katodritou:Theagenion Cancer Hospital: Current Employment; Karyopharm: Research Funding; Abbvie: Research Funding; Genesis Pharma: Honoraria, Other: Expenses, Research Funding; Takeda: Honoraria, Other: Expenses, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen-Cilag: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Verrou:Abbvie: Honoraria; Amgen: Honoraria; Genesis: Honoraria; Roche: Honoraria; Takeda: Honoraria; Karyopharm: Research Funding; Janssen Cilag: Honoraria, Research Funding. Gavriatopoulou:Amgen: Consultancy, Honoraria; Karyopharm: Consultancy, Honoraria; Genesis Pharma: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Takeda: Consultancy, Honoraria. Leonidakis:Health Data Specialists S.A.: Current Employment. Delimpasi:Janssen: Honoraria; GENESIS: Honoraria; Takeda: Honoraria; Amgen: Honoraria. Kyrtsonis:Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Genesis pharma SA: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Papaioannou:Gilead: Honoraria; Genesis Pharma: Honoraria; Janssen-Cilag: Honoraria; Alexion: Membership on an entity's Board of Directors or advisory committees. Symeonidis:Sanofi/Genzyme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; WinMedica: Research Funding; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Research Funding; Astellas: Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bristol-Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; GenesisPharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck Sharp & Dohme: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Dimopoulos:Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Speakers Bureau; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees, Research Funding, Speakers Bureau; BMS: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Personal fees.
- Published
- 2020
- Full Text
- View/download PDF
33. A revised international prognostic score system for Waldenström's macroglobulinemia
- Author
-
Efstathios, Kastritis, Pierre, Morel, Alain, Duhamel, Maria, Gavriatopoulou, Marie Christine, Kyrtsonis, Eric, Durot, Argiris, Symeonidis, Kamel, Laribi, Evdoxia, Hatjiharissi, Loic, Ysebaert, Amalia, Vassou, Nikolaos, Giannakoulas, Giampaolo, Merlini, Panagiotis, Repousis, Marzia, Varettoni, Euridyki, Michalis, Bénédicte, Hivert, Michalis, Michail, Eirini, Katodritou, Evangelos, Terpos, Veronique, Leblond, and Meletios A, Dimopoulos
- Subjects
Adult ,Aged, 80 and over ,International Cooperation ,Hematology ,Middle Aged ,Medical Oncology ,Prognosis ,Risk Assessment ,Severity of Illness Index ,Survival Rate ,Recurrence ,Humans ,Immunotherapy ,Prospective Studies ,Waldenstrom Macroglobulinemia ,Rituximab ,Aged ,Follow-Up Studies ,Neoplasm Staging ,Proportional Hazards Models - Abstract
A staging system was developed a decade ago for patients with Waldenström's macroglobulinemia (WM), however, since then WM treatments have changed. A revised staging system could better capture prognosis of WM patients in the chemoimmunotherapy era. We developed a revised system based on data from 492 symptomatic patients with at least 3 years and a median of 7 years of follow up while an independent validation cohort included 229 symptomatic patients. We identified age (≤65 vs 66-75 vs ≥76 years), b2-microglobulin ≥ 4 mg/L, serum albumin3.5 gr/dl, and LDH ≥ 250 IU/L (ULN 225) to stratify patients in five different prognostic groups and identify a very-low risk as well as a very-high risk group with a 3-year WM-related death rate of 0, 10, 14, 38, and 48% (p 0.001) and 10-year survival rate of 84, 59, 37, 19, and 9% (p 0.001). We evaluated this staging system separately in patients65 years and65 years, according to the reason for initiation of treatment, among patients receiving frontline rituximab or in patients treated primarily without rituximab. With further validation before clinical use, this revised IPSSWM could improve WM patient risk stratification, is easily available and may be used in the everyday practice to provide prognostic information.
- Published
- 2018
34. The BCL2 antagonist ABT-199 triggers apoptosis, and augments ibrutinib and idelalisib mediated cytotoxicity inCXCR4Wild-typeandCXCR4WHIMmutated Waldenstrom macroglobulinaemia cells
- Author
-
Matthew S. Davids, Jie Chen, Steven P. Treon, Guang Yang, Nickolas Tsakmaklis, Yang Cao, Sandra Kanan, Xia Liu, Jorge J. Castillo, Evdoxia Hatjiharissi, Zachary R. Hunter, and Lian Xu
- Subjects
Receptors, CXCR4 ,Apoptosis ,CXCR4 ,chemistry.chemical_compound ,Piperidines ,Proto-Oncogene Proteins ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Receptor ,Cytotoxicity ,Quinazolinones ,Sulfonamides ,Bcl-2-Like Protein 11 ,business.industry ,Adenine ,Wild type ,Membrane Proteins ,Waldenstrom macroglobulinemia ,Drug Synergism ,Hematology ,Bridged Bicyclo Compounds, Heterocyclic ,medicine.disease ,Pyrimidines ,chemistry ,Purines ,Ibrutinib ,Cancer research ,Pyrazoles ,Waldenstrom Macroglobulinemia ,Apoptosis Regulatory Proteins ,business ,Idelalisib - Published
- 2015
- Full Text
- View/download PDF
35. Efficacy of Daratumumab with Dexamethasone in Patients with Relapsed/Refractory Multiple Myeloma and Severe Renal Impairment: An Interim Analysis of a Phase 2 Study (the DARE Study)
- Author
-
Evangelos Terpos, Despina Fotiou, Eftathios Kastritis, Elena Zamagni, Meletios A. Dimopoulos, Maria Gavriatopoulou, Nikolaos Kanellias, Marie-Christine Kyrtsonis, Barbara Gamberi, Magdalini Migkou, Michele Cavo, Argiris Symeonidis, Eirini Katodritou, Maria Roussou, Evdoxia Hatjiharissi, and Sosana Delimpasi
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Bortezomib ,medicine.medical_treatment ,Immunology ,Daratumumab ,Phases of clinical research ,Cell Biology ,Hematology ,Interim analysis ,medicine.disease ,Biochemistry ,Internal medicine ,Medicine ,Hemodialysis ,business ,health care economics and organizations ,Dexamethasone ,Multiple myeloma ,Lenalidomide ,medicine.drug - Abstract
Introduction: About 20-40% of patients (pts) with multiple myeloma (MM) present with moderate or severe renal impairment (RI) and about 25% of pts will also experience RI later during the disease course (Dimopoulos MA, et al; Leukemia 2008; 22:1485-1493). Moderate or severe RI is associated with poorer overall survival (OS) and higher risk of early death but also with challenges in the management and administration of the appropriate treatment. Daratumumab, an IgG1κ human monoclonal antibody that targets CD38, has shown efficacy and a favorable safety profile in pts with relapsed or refractory MM (RRMM) both as a monotherapy (Lonial S, et al; Lancet 2016; 387(10027):1551-1560) and in combination with other anti-myeloma agents (Dimopoulos MA, et al; N Engl J Med 2016; 375:1319-1331, and Palumbo A, et al; N Engl J Med 2016; 375:754-766). In population pharmacokinetic analyses, no clinically important differences in exposure to daratumumab were observed between pts with renal impairment and those with normal renal function. However, there are no prospective data on the safety and efficacy of daratumumab in pts with RRMM and severe renal dysfunction or those requiring dialysis. Methods: DARE is an ongoing multicenter, single arm, open-label, phase 2 study, aiming to enroll ~38 adult pts with documented RRMM and severe renal impairment (estimated glomerular filtration rate [eGFR]< 30 ml/min/1.73m2) or in need for hemodialysis. Pts must previously have had ≥ 2 lines of therapy with both bortezomib- and lenalidomide-based regimens and an Eastern Cooperative Oncology Group performance status (ECOG PS) score of ≤ 2. Exclusion criteria include previous treatment with daratumumab or other anti-CD38 therapy. All pts receive 28-day cycles of treatment with daratumumab given intravenously at 16 mg/kg weekly for Cycles 1-2, every 2 weeks for Cycles 3-6, followed by every 4 weeks thereafter, with dexamethasone 40 mg given weekly for each 4-week cycle. The primary endpoint of the study is the evaluation of progression-free survival (PFS). Key secondary endpoints include overall response rate (ORR; defined as the proportion of pts with partial response or better), renal response rate (RRR; defined as the proportion of pts with best response of renal partial response or better), and the assessment of daratumumab safety and tolerability. All responses were based on investigators' assessment per International Myeloma Working Group criteria. This interim analysis presents study results for pts who received the first dose of study treatment at least 3 months prior to the cut-off date (06/05/2019). Results: Eighteen pts, enrolled in 4 centers, were included in this prespecified analysis. The pts' median age was 74.0 years, and most were males (77.8%). The median time from MM diagnosis to first dose of daratumumab was 3.6 years. The number of pts with baseline ECOG PS 0, 1, and 2 were 4 (22.2%), 13 (72.2%), and 1 (5.6%), respectively. At the start of the study, 22.2% and 77.8% of patients had ISS Stage II and III disease, respectively. Moreover, 44.4%, and 55.6% of pts had a revised ISS stage II and III, respectively. Median number of prior lines of therapy was 3.5, and two (11.1%) pts had previous autologous stem cell transplantation; Median eGFR at baseline was 12 mL/min/1.73m2. The median number of therapy cycles received per patient was 4.5. The median time from first daratumumab dose to first partial response or better was 0.9 months. The median follow-up is 4.4 months and the Kaplan-Meier estimate of the 6-month PFS rate is 51.9% (Figure), ORR was 44.4% (8/18 pts) (including VGPR in 4, and PR in 4 pts), and RRR was 27.8% (5/18 pts). By the cut-off date, 10 (55.6%) pts were still on daratumumab; 7 (38.9%) pts discontinued treatment due to progressive disease and 1 (5.6%) due to a fatal serious adverse event (SAE). Overall, ten (55.6%) pts had ≥ 1 AE of grade 3 or 4. Of all grade 3 or 4 AEs, the most frequent were anemia (4, 25%), hyperglycemia (3, 18.8%), and hypocalcemia (2, 12.5%). Three (16.7%) pts suffered a single SAE each: lung infection, sepsis (fatal), and stroke. Conclusions: The combination of daratumumab with dexamethasone is efficacious and with a favorable safety profile and no new safety signals, in pts with RRMM with severe renal impairment. Importantly, hematologic responses are high in these heavily pretreated patients while about 28% achieved also a renal response. The study is ongoing and updated results will be presented at the meeting. Figure Disclosures Kastritis: Pfizer: Honoraria; Prothena: Honoraria; Genesis: Honoraria; Amgen: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Takeda: Honoraria. Terpos:Takeda: Honoraria, Other: Travel expenses, Research Funding; Genesis: Honoraria, Other: Travel expenses, Research Funding; Celgene: Honoraria; Medison: Honoraria; Amgen: Honoraria, Research Funding; Janssen: Honoraria, Other: Travel expenses, Research Funding. Symeonidis:Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Research Funding; Tekeda: Membership on an entity's Board of Directors or advisory committees, Research Funding; MSD: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding. Delimpasi:Genesis: Honoraria, Other: Travel grant; Janssen: Honoraria; Takeda: Honoraria; Amgen: Honoraria. Cavo:Janssen, Celgene: Other: Travel Accommodations; Janssen, Celgene, Amgen, Abbvie: Consultancy, Membership on an entity's Board of Directors or advisory committees; Janssen, Celgene: Speakers Bureau; Celgene, Janssen, Amgen, BMS, Abbvie, Takeda: Honoraria. Zamagni:Celgene Corporation: Honoraria, Other: Advisory board, Speakers Bureau; Janssen: Honoraria, Other: Advisory board, Speakers Bureau; Amgen: Honoraria, Other: Advisory board, Speakers Bureau; BMS: Honoraria, Other: Advisory Board, Speakers Bureau; Takeda: Honoraria, Speakers Bureau; Sanofi: Honoraria, Other: Advisory Board, Speakers Bureau. Katodritou:Takeda: Honoraria; Janssen: Honoraria; Genesis: Honoraria; Amgen: Honoraria. Gamberi:Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Gavriatopoulou:Genesis: Honoraria, Other: Travel expenses; Amgen: Honoraria; Janssen: Honoraria, Other: Travel expenses; Takeda: Honoraria, Other: Travel expenses. Hatjiharissi:Janssen: Honoraria. Dimopoulos:Sanofi Oncology: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
36. Oncogenic MAF in Co-Operation with IRF4 Confers Extensive Chromatin Re-Arrangement in Plasma Cells and Generates 'Neo-Enhancers' That Regulate Genes Critical for Myeloma Biology
- Author
-
Alexia Katsarou, Ian Sudbery, Valentina S. Caputo, Foteini Papaleonidopoulou, Kanagaraju Ponnusamy, Aristeidis Chaidos, Maria Papaioannou, Niklas Feldhahn, Nikolaos Trasanidis, Xiaolin Xiao, Anastasios Karadimitris, Keren Keren, Jaime Alvarez-Benayas, Irene Roberts, Ioannis Kostopoulos, and Evdoxia Hatjiharissi
- Subjects
Chromatin binding ,Immunology ,NUAK1 ,Promoter ,Cell Biology ,Hematology ,Transcription Factor Maf ,Biology ,Biochemistry ,Cell biology ,Chromatin ,Cistrome ,Enhancer ,Epigenomics - Abstract
Overexpression of the transcription factor MAF, as a result of its juxtaposition to the IgH enhancer [MAF-translocated t(14;16)], is a myeloma-initiating event in 3-5% of patients with multiple myeloma (MM) and confers a poor prognosis. MAF is also overexpressed in another 40% of cases, often in co-operation with the oncogene MMSET. The mechanisms by which MAF overexpression impacts on the regulatory genome to generate the MAF-driven oncogenic transcriptome and its direct targets are not known. To address this, we employed a multi-layer -omics approach using primary myeloma plasma cells (PC) as well as myeloma cell lines (MMCL). First, we determined the chromatin accessibility and transcriptome profiles of MAF-translocated myeloma by performing ATAC-seq and RNA-seq, respectively, in purified bone marrow CD138+ PC from two patients with t(14;16) and three healthy donors. We identified 6,640 differentially accessible regions, 87% of which displayed enhanced chromatin accessibility in MAF samples compared to normal PC. Secondary analysis comparing this with ATAC-seq data from a set of 28 other MM samples, including hyperdiploid, MMSET and CCND1-translocated MM, revealed 33% of those regions to be MAF subgroup specific (1,949 regions), with the rest shared between MAF and other cytogenetic groups. Gene annotation and pathway enrichment analysis using GREAT confirmed overrepresentation of the MF myeloma patient signature, as previously identified in microarray datasets. RNA-seq detected significant upregulation of approximately 900 genes in MAF samples compared to normal counterparts, including MAF itself (top 4th hit) as well as its presumed targets (CCND2, ITGB7 and NUAK1). Next, we obtained the MAF cistrome using ChIP-seq in the MAF-translocated MMCL MM1.S and integrated it with the primary PC ATAC-seq data. This revealed that 31% (618/1,949) of the differentially accessible regions in MAF-translocated MM PC are also MAF-bound. Additional overlay with ENCODE ChromHMM epigenome map showed that 47% of MAF binding sites are on active enhancers and 42% on active promoters signifying potential direct regulation of the corresponding genes. Next, we superimposed the accessible and MAF-bound loci on the epigenomic landscapes of normal PC and other B-cell types using their corresponding ChromHMM maps (Blueprint consortium data). Interestingly, 56% (345/618) of the MAF-specific regions were not active in any stage of B cell development. This suggests that aberrant MAF overexpression and chromatin binding in PC is associated with de novo activation of these chromatin regions, over half of which (200/345; 58%) are enhancers; we termed these 'neo-enhancers'. Upon de novo motif analysis of MAF ChIP-seq in MAF-translocated JJN3 and MM1.S MMCL, we confirmed MAF as the first and, interestingly, IRF4 as the second top hit, suggesting a possible MAF-IRF4 functional interaction in myelomagenesis. Indeed, overlay of the accessible MAF-bound loci with IRF4 ChIP-seq data in MM1.S revealed 63% co-occupancy (including 62% of "neo-enhancers"), proposing a novel and extensive co-operative chromatin-based network between the two transcription factors. Final integration of the accessible MAF-bound regions with the paired transcriptomes of primary myeloma PC revealed a set 206 candidate enhancer-gene pairs. Strikingly, we identified two IRF4-cobound "neo-enhancers" linked to overexpression of TLR4 and CCR1, two genes known for their roles in myeloma cell proliferation and migration. We confirmed significant downregulation of both genes upon shRNA-mediated knockdown of MAF in the two MAF-translocated MMCL, MM1.S and JJN3, as well as the lethality of MAF depletion. Further, MAF overexpression in MAF-negative myeloma backgrounds led to transcriptional upregulation of these genes, further validating them as MAF targets. While CRISPR/Cas9i experiments targeting TLR4 are ongoing, preliminary results validated the functional role of the "neo-enhancer" in CCR1 gene expression. In conclusion, we demonstrate for the first time an extensive re-organisation of the PC chromatin conferred by oncogenic MAF in MM; we reveal its extensive co-operation with IRF4 in this process; we validate the directly MAF-regulated genes and functionally characterise neo-enhancers of key MAF-dependent genes that in addition to MAF itself are also critical for myeloma biology. Disclosures Hatjiharissi: Janssen: Honoraria. Caputo:GSK: Research Funding. Karadimitris:GSK: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
37. Longitudinal T Cell Immunoprofiling of Patients with Relapsed and/or Refractory Myeloma Who Receive Daratumumab Monotherapy: A Subanalysis of a Phase 2 Study (the REBUILD Study)
- Author
-
Konstantia Kotta, Argiris Symeonidis, Electra Sofou, Eftathios Kastritis, Sosana Delimpasi, Anna Vardi, Anastasia Chatzidimitriou, Eirini Katodritou, Maria Gavriatopoulou, Alexandra Siorenta, Evangelos Terpos, Maria Karipidou, Meletios A. Dimopoulos, Evdoxia Hatjiharissi, Elisavet Vlachonikola, and Kostas Stamatopoulos
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,T cell ,Immunology ,Phases of clinical research ,Daratumumab ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,medicine.anatomical_structure ,Refractory ,Internal medicine ,Partial response ,medicine ,business ,Multiple myeloma ,Lenalidomide ,medicine.drug - Abstract
Recent evidence suggests immunomodulatory effects of daratumumab in heavily pre-treated Multiple Myeloma (MM) patients (pts); however, the precise effects remain under-characterized, particularly at the molecular level. REBUILD is an ongoing prospective, multicenter, non-comparative, open-label, phase II study that evaluates the effects of daratumumab monotherapy on bone metabolism of pts with relapsed and/or refractory MM (RRMM) who have had ≥2 prior lines of therapy, including lenalidomide and a proteasome inhibitor. Secondary endpoint of the study included the evaluation of T cell dynamics by comprehensive analysis of the T cell receptor (TR) repertoire employing next generation sequencing (NGS) and multi-color flow cytometry. Herein we report the results of this secondary endpoint for the first 14 pts who completed 3 cycles of daratumumab monotherapy. In total, we analyzed 28 paired samples collected at screening (n=14) and on Day 1 of Cycle 4 (C4D1, n=14) of treatment in order to assess potential changes in relation to treatment and clinical response. Patients were grouped based on best responses into responders (i.e. patients with partial response [PR, n=1] and very good PR [VGPR, n=6]), and non-responders (i.e. patients with minimal response [MR, n=2], stable disease [SD, n=4], or progressive disease [PD, n=1]). Starting material was peripheral blood mononuclear cells. TRBV-TRBD-TRBJ gene rearrangements were RT-PCR amplified and subjected to paired-end NGS. Raw NGS reads (n=6,715,406 | median 221,145/sample) were processed through a previously published, purpose-built bioinformatics pipeline. Only productive TRBV-TRBD-TRBJ rearrangements were taken into consideration (n=3,097,565 | median 101,670/sample) for the computation of clonotypes (i.e. TRB rearrangements with identical TRBV gene usage and amino acid complementarity-determining region 3 sequence). Overall, 151,153 distinct clonotypes (median 5,084 clonotypes/sample) were assessed. Both groups (responders/non-responders) displayed clonal T cell expansions both pre- and post-treatment. Clonality was found to be increased after treatment for both responders and non-responders, with statistical significance in the former (median cumulative frequency of the 10 most expanded T cell clonotypes/sample: 31% pre-treatment versus 40% post-treatment, respectively | p=0.04). In both groups, the clonotype repertoire appeared to be renewed with only a small fraction of pre-treatment clonotypes remaining after treatment (1% for non-responders; 0.6% for responders). Interestingly, in the responders' group we noticed a significant shift in the major clonotype repertoire at screening vs C4D1. In particular, in the responders' group the 10 most expanded clonotypes/sample at C4D1 represented expansions of clonotypes present at very low frequency at screening, whereas the most expanded clonotypes at screening decreased or even diminished post-treatment, suggesting that daratumumab treatment led to the emergence of anti-myeloma T cell clones which contributed to clinical response. On the contrary, the 10 most expanded pre-treatment clonotypes in the non-responders' group tended to dominate also the post-treatment repertoire. Of note, 13 shared clonotypes were identified amongst the post-treatment repertoires of different patients (responders/non-responders); shared clonotypes were not found in other entities in public databases, raising the possibility that they may be "disease-specific" and selected by common tumor-associated antigens. With a single exception, shared clonotypes were detected in cases with relevant HLA restrictions, which is noteworthy given the random HLA background of our cohort. Finally, flow cytometry analysis revealed a significant increase post treatment in the percentage of CD3+ T cells (median frequency at screening 60% versus 83% at C4D1 | p=0.003), driven mostly by the expansion of the CD8+ T cell compartment (median frequency at screening 30.8% versus 48.9% at C4D1 | p=0.03) in both groups. In conclusion, TR clonality increases post-treatment through a renewal mechanism; however, pre-treatment clones significantly expanded post-treatment in responders, alluding to the existence of clonotypes with anti-MM properties that may be activated after treatment with daratumumab, arguably contributing to clinical response. Disclosures Kastritis: Janssen: Honoraria, Research Funding; Takeda: Honoraria; Pfizer: Honoraria; Prothena: Honoraria; Genesis: Honoraria; Amgen: Honoraria, Research Funding. Hatjiharissi:Janssen: Honoraria. Katodritou:Genesis: Honoraria; Janssen: Honoraria; Takeda: Honoraria; Amgen: Honoraria. Gavriatopoulou:Genesis: Honoraria, Other: Travel expenses; Janssen: Honoraria, Other: Travel expenses; Takeda: Honoraria, Other: Travel expenses; Amgen: Honoraria. Delimpasi:Takeda: Honoraria; Amgen: Honoraria; Janssen: Honoraria; Genesis: Honoraria, Other: Travel grant. Symeonidis:Sanofi: Research Funding; MSD: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; Tekeda: Membership on an entity's Board of Directors or advisory committees, Research Funding. Stamatopoulos:Janssen: Honoraria, Research Funding; Abbvie: Honoraria, Research Funding. Dimopoulos:Sanofi Oncology: Research Funding. Terpos:Janssen: Honoraria, Other: Travel expenses, Research Funding; Medison: Honoraria; Genesis: Honoraria, Other: Travel expenses, Research Funding; Amgen: Honoraria, Research Funding; Celgene: Honoraria; Takeda: Honoraria, Other: Travel expenses, Research Funding. Chatzidimitriou:Janssen: Honoraria.
- Published
- 2019
- Full Text
- View/download PDF
38. Impact of Daratumumab Monotherapy on Bone Parameters in Patients with Relapsed and/or Refractory Multiple Myeloma Who Have Received at Least 2 Prior Lines of Therapy Including Lenalidomide and a Proteasome Inhibitor; Interim Analysis of a Phase 2 Study (the REBUILD Study)
- Author
-
Eirini Katodritou, Evdoxia Hatjiharissi, Alexandros Leonidakis, Eftathios Kastritis, Evgenia Verrou, Sosana Delimpasi, Evangelos Terpos, Argiris Symeonidis, Maria Gavriatopoulou, Marie-Christine Kyrtsonis, Meletios A. Dimopoulos, Evangelos Eleutherakis-Papaiakovou, Maria Papaioannou, and Ioannis Ntanasis-Stathopoulos
- Subjects
Oncology ,medicine.medical_specialty ,education.field_of_study ,Bone disease ,business.industry ,Surrogate endpoint ,Immunology ,Population ,Daratumumab ,Cell Biology ,Hematology ,medicine.disease ,Interim analysis ,Biochemistry ,N-terminal telopeptide ,Internal medicine ,medicine ,Clinical endpoint ,business ,education ,Lenalidomide ,medicine.drug - Abstract
Introduction: A major clinical feature of multiple myeloma (MM) is osteolytic bone disease, with bone resorption prevailing bone formation. Daratumumab, an IgG1κ human monoclonal antibody that targets CD38, has been licensed for use in patients (pts) with relapsed or refractory MM (RRMM); however, its effect on bone disease has not yet been determined in a clinical setting. CD38 expression has been associated with osteoclast formation and bone resorption in in vitro studies and treatment with daratumumab has demonstrated an inhibitory effect on osteoclastogenesis. This study aims to evaluate the impact of daratumumab monotherapy on biochemical markers of bone metabolism in RRMM pts. Methods: REBUILD is an ongoing prospective, multicenter, non-comparative, open-label, phase II study aiming to enroll 57 adult pts with documented RRMM who have had ≥2 prior lines of therapy, including lenalidomide and a proteasome inhibitor. Pts should have a Karnofsky Performance Status score of ≥70, and a creatinine clearance of ≥30 mL/min. Exclusion criteria include previous treatment with daratumumab or other anti-CD38 therapy. Pts receive daratumumab at a weekly dose of 16 mg/kg for Cycles 1-2, every 2 weeks for Cycles 3-6 and every 4 weeks thereafter. The primary endpoint of this study is the change from baseline in the bone resorption markers C-terminal telopeptide of collagen type I (CTX) and tartrate-resistant acid phosphatase isoform 5b (TRACP-5b) after 4 months of daratumumab monotherapy. Secondary endpoints include the change at 4 months from baseline in bone formation markers (bone-specific alkaline phosphatase [bALP], osteocalcin [OC], and procollagen type-I N-propeptide [PINP]), in markers of osteoclast regulation (receptor activator of nuclear factor kappa-B ligand [RANKL], osteoprotegerin [OPG] and chemokine (C-C motif) ligand-3 [CCL-3]), osteoblast control (sclerostin, dickkopf-1 [DKK-1]), and progression-free survival (PFS). This interim analysis included pts who received the first dose of study treatment at least 6 months before the cut-off date (26/04/2019). Results: Twenty-seven pts had been enrolled in 5 study centres of the Greek Myeloma Study Group, and among them 15 had both biomarker and clinical data available after 4 months of therapy and they were included in the present analyses. The median number of previous therapies was 3. Nine (60%) pts had >10 lytic bone lesions at screening. Only 3/15 (20%) pts received bisphosphonates along with daratumumab monotherapy. Regarding the primary endpoint, the median change of CTX and TRACP-5b levels at 4 months from baseline was 13.4%, and -2.6%, respectively. The median change in CTX and TRACP-5b levels at 4 months from baseline for pts with partial response or better (≥PR) (n=7, 46.7%) was 13.4% and -9.4%, respectively; among pts with minimal response or stable disease (n=6, 40%) the change in CTX and TRACP-5b levels was 16.4% and 14.3%, respectively. The levels of all bone formation markers bALP, OC, and PINP showed a median increase at 4 months from baseline by 18.4%, 190.5%, and 19.3%, respectively. For pts with ≥PR, the median change at 4 months from baseline in bALP, OC, and PINP levels was even higher: 25.3%, 338.7%, and 19.3%, respectively. Interestingly, the median change at 4 months from baseline in osteoblast inhibitors sclerostin and DKK-1 levels was -20.3%, and -6.5%, respectively. The median PFS for all 27 pts enrolled was 7.1 months. Conclusions: Regarding the primary endpoint of the REBUILD study, results from this interim analysis indicate that daratumumab monotherapy decreases TRACP-5b levels, which was more pronounced in responders, but this is not accompanied by a reduction in CTX. An effect on CTX levels may become evident with additional follow-up. However, a clear increase in serum levels of all markers of bone formation tested (bALP, OC, PINP) was observed, especially among responders, which might at least partially be explained by a reduction of the osteoblast inhibitors sclerostin and DKK-1. Thus, daratumumab monotherapy in this RRMM population (heavily pre-treated pts with multiple lytic bone lesions and limited use of bisphosphonates) results in a decrease in TRACP-5b and osteoblast inhibitors and an increase in bone formation markers, suggesting an overall positive impact on bone remodeling. The study is ongoing and updated results will be presented at the meeting. Disclosures Terpos: Takeda: Honoraria, Other: Travel expenses, Research Funding; Genesis: Honoraria, Other: Travel expenses, Research Funding; Janssen: Honoraria, Other: Travel expenses, Research Funding; Amgen: Honoraria, Research Funding; Celgene: Honoraria; Medison: Honoraria. Kastritis:Amgen: Honoraria, Research Funding; Pfizer: Honoraria; Janssen: Honoraria, Research Funding; Takeda: Honoraria; Prothena: Honoraria; Genesis: Honoraria. Hatjiharissi:Janssen: Honoraria. Katodritou:Takeda: Honoraria; Janssen: Honoraria; Genesis: Honoraria; Amgen: Honoraria. Gavriatopoulou:Takeda: Honoraria, Other: Travel expenses; Genesis: Honoraria, Other: Travel expenses; Janssen: Honoraria, Other: Travel expenses; Amgen: Honoraria. Leonidakis:HeaDS: Employment. Delimpasi:Janssen: Honoraria; Genesis: Honoraria, Other: Travel grant; Amgen: Honoraria; Takeda: Honoraria. Symeonidis:MSD: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Research Funding; Tekeda: Membership on an entity's Board of Directors or advisory committees, Research Funding. Dimopoulos:Sanofi Oncology: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
39. Distinct Chromatin Accessibility Changes, Aberrant Transcription Factor Networks Combined with Novel Oncogenic Enhancers Characterise Myeloma-Initiating Genetic Events
- Author
-
Maria Papaioannou, Nikolaos Trasanidis, Ian Sudbery, Philippa C. May, Xiaolin Xiao, Irene Roberts, Anastasios Karadimitris, Kanagaraju Ponnusamy, Aristeidis Chaidos, Valentina S. Caputo, Marco Bua, Evdoxia Hatjiharissi, Maria Atta, Alexia Katsarou, Holger W. Auner, and Jaime Alvarez-Benayas
- Subjects
biology ,Immunology ,RNA ,Chromosomal translocation ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Chromatin ,Histone ,Cyclin D1 ,Cancer research ,biology.protein ,medicine ,Precordial catch syndrome ,Enhancer ,Transcription factor - Abstract
In multiple myeloma (MM), a malignancy of the bone marrow plasma cells (BMPC), hyperdiploidy (HY) and oncogene over-expression via chromosomal translocation [including CCND1- t(11;14), MAF- t(14;16), MMSET-t(4;14)] are the primary myeloma initiating events (MIE) that drive distinct transcriptional programs. These are further shaped by secondary SNV and CNV events. This genetic heterogeneity converges, in most cases, to a functionally dichotomous state of CCND1 or CCND2 overexpression. The molecular mechanisms underlying each of the distinct myelomagenic transcriptomes and the CCND1 vs CCND2 dichotomy have not been defined. To address these questions, we obtained highly purified BMPC from 3 healthy donors and 30 MM patients (HY: 15; CCND1: 4; MMSET: 5; MAF: 2; other: 4), either at diagnosis or relapse, and mapped their chromatin accessibility and transcriptome profiles by ATAC-seq and RNA-seq, respectively. In total, we obtained ~300K regions with accessible chromatin in either MM or normal PC. Overall chromatin accessibility increased in myeloma compared to normal PC, particularly in MAF- and MMSET-translocated subtypes. Analysis of combined ATAC-seq/RNA-seq by Multi-Omics Factor Analysis (MOFA) resulted in a clearer samples distinction than either ATAC-seq or RNA-seq alone, with altered chromatin accessibility accounting for more of the variance than expression. Of the top five identified factors, the top two (one transcriptome driven, one accessibility driven) distinguished normal from MM samples, whilst two more separated MMSET, MAF and CCND1 subgroups. Ninety seven, 157, 256 and 348 overexpressed genes in the CCND1, HY, MMSET and MAF subgroups, respectively, were predicted to be regulated by differentially accessible enhancers. Twenty percent (165/858) of these genes were overexpressed in >1 subgroup suggesting a process of chromatin accessibility-based convergence evolution. Enrichment analysis suggested direct or indirect involvement of Polycomb and chromatin remodellers; significant enrichment was also found for genes involved in neurogenesis. ATAC-seq footprinting predicted binding sites for 250 expressed transcription factors (TFs), 116 of which displayed higher binding frequency in myeloma than in normal PC and included both known (e.g., XBP1, RELA, IRF4, PRDM1) and potentially novel regulators of myeloma biology (e.g., CXXC1 and NFE2L1). The remaining 134 TF were predicted to be present in at least one MM subgroup, but absent in normal PC. Amongst them, as expected, MAF was active in the MMSET- and more so in the MAF-translocated subgroups. DepMap database analysis suggested myeloma cell dependency on 181/250 TF (CRISPR/Cas9 CERES score < -0.1 in >3/14 MMCL analysed). In dissecting the regulatory basis of CCND2 vs CCND1 dichotomy, one MOFA factor completely separated MAF from CCND1 samples, placing extreme opposite weights on the expression of CCND2 and CCND1 respectively. Interestingly, the same factor identified open-chromatin clusters upstream of CCND2 and linked them to its over-expression. These clusters were also open in the MMSET group and in CCND2-expressing HY samples. Conversely, no accessibility was detected in the CCND1 group, the CCND1-expressing HY samples or in normal PCs. Further, super-enhancer calling using the H3K27ac histone mark in MAF-translocated JJN3 cells identified the region of interest as a bona fide super-enhancer. Chromatin long range interactions, as assessed by Capture-HiC, demonstrated high frequency interactions of the CCND2 promoter with the constituent elements of the putative super-enhancer. Experimental validation using a CRISPR/Cas9i system confirmed the functional role of all 4 super-enhancer constituents tested in the regulation of CCND2 expression, while TF footprinting predicted MAF binding to the super-enhancer in MAF-translocated PC. In conclusion, we show that distinct oncogenic transcriptomes in MM are underpinned by extensive chromatin changes, accompanied by TF activity 're-wiring' that does not necessarily require transcriptional deregulation of the TF themselves. We identify novel, non-oncogene TF dependencies that suggest therapeutic opportunities in MM and we discover and characterise the critical super-enhancer that drives overexpression of the CCND2 oncogene in MM. Disclosures Auner: Amgen: Other: Consultancy and Research Funding; Takeda: Consultancy; Karyopharm: Consultancy. Hatjiharissi:Janssen: Honoraria. Caputo:GSK: Research Funding. Karadimitris:GSK: Research Funding.
- Published
- 2019
- Full Text
- View/download PDF
40. Glycosphingolipid synthesis inhibition limits osteoclast activation and myeloma bone disease
- Author
-
Raymond A. Dwek, Lynett Danks, Simon Parry, Aristeidis Chaidos, Emmanouil Spanoudakis, Adel Ersek, Anastasios Karadimitris, Gabriele Twigg, Evdoxia Hatjiharissi, Terry D. Butters, Aristotelis Antonopoulos, Irene Roberts, Amin Rahemtulla, Ming Hu, A Freidin, Stuart M. Haslam, Youridies Vattakuzhi, Anne Dell, Lynn M. Williams, Ke Xu, Nicole J. Horwood, Katerina Goudevenou, Dominic S. Alonzi, Ana Isabel Espirito Santo, Maria Papaioannou, and Biotechnology and Biological Sciences Research Council (BBSRC)
- Subjects
CELL LINE HL-60 ,Osteolysis ,medicine.medical_treatment ,Cell ,MACROPHAGE INFLAMMATORY PROTEIN-1-ALPHA ,N-BUTYLDEOXYNOJIRIMYCIN ,Osteoclasts ,Research & Experimental Medicine ,GAUCHER-DISEASE ,CSK Tyrosine-Protein Kinase ,PROTEIN 1-ALPHA ,Mice ,Insulin-Like Growth Factor I ,Lipid raft ,Mice, Knockout ,biology ,General Medicine ,11 Medical And Health Sciences ,medicine.anatomical_structure ,KAPPA-B LIGAND ,src-Family Kinases ,Medicine, Research & Experimental ,RANKL ,Glucosyltransferases ,lipids (amino acids, peptides, and proteins) ,Female ,Signal transduction ,Multiple Myeloma ,Life Sciences & Biomedicine ,RECEPTOR ACTIVATOR ,Research Article ,medicine.medical_specialty ,1-Deoxynojirimycin ,Immunology ,Plasma cell dyscrasia ,SIGNAL-TRANSDUCTION ,Glycosphingolipids ,Cell Line ,Membrane Microdomains ,Osteoclast ,Internal medicine ,MULTIPLE-MYELOMA ,medicine ,Animals ,Glycoside Hydrolase Inhibitors ,TNF Receptor-Associated Factor 6 ,Science & Technology ,Growth factor ,RANK Ligand ,medicine.disease ,ALPHA MIP-1-ALPHA ,Endocrinology ,biology.protein ,Cancer research - Abstract
Glycosphingolipids (GSLs) are essential constituents of cell membranes and lipid rafts and can modulate signal transduction events. The contribution of GSLs in osteoclast (OC) activation and osteolytic bone diseases in malignancies such as the plasma cell dyscrasia multiple myeloma (MM) is not known. Here, we tested the hypothesis that pathological activation of OCs in MM requires de novo GSL synthesis and is further enhanced by myeloma cell-derived GSLs. Glucosylceramide synthase (GCS) inhibitors, including the clinically approved agent N-butyl-deoxynojirimycin (NB-DNJ), prevented OC development and activation by disrupting RANKL-induced localization of TRAF6 and c-SRC into lipid rafts and preventing nuclear accumulation of transcriptional activator NFATc1. GM3 was the prevailing GSL produced by patient-derived myeloma cells and MM cell lines, and exogenous addition of GM3 synergistically enhanced the ability of the pro-osteoclastogenic factors RANKL and insulin-like growth factor 1 (IGF-1) to induce osteoclastogenesis in precursors. In WT mice, administration of GM3 increased OC numbers and activity, an effect that was reversed by treatment with NB-DNJ. In a murine MM model, treatment with NB-DNJ markedly improved osteolytic bone disease symptoms. Together, these data demonstrate that both tumor-derived and de novo synthesized GSLs influence osteoclastogenesis and suggest that NB-DNJ may reduce pathological OC activation and bone destruction associated with MM.
- Published
- 2016
- Full Text
- View/download PDF
41. A Revised Staging System for Waldenström's Macroglobulinemia
- Author
-
Marie-Christine Kyrtsonis, Evangelos Terpos, Michalis Michael, Panagiotis Panagiotidis, Nikolaos Giannakoulas, Stavroula Giannouli, Panagiotis Repousis, Emmanouil Spanoudakis, Eirini Katodritou, Argiris Symeonidis, Evdoxia Hatjiharissi, Efstathios Kastritis, Aikaterini Megalakaki, Anastasia Pouli, Maria Gavriatopoulou, Dimitrios Christoulas, Panagiotis Tsirigotis, Elina Vervessou, and Meletios-Athanasios Dimopoulos
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,medicine ,Macroglobulinemia ,Hematology ,Radiology ,business ,Staging system - Published
- 2017
- Full Text
- View/download PDF
42. Histone Deacetylase Inhibitors Demonstrate Significant Preclinical Activity as Single Agents, and in Combination with Bortezomib in Waldenström's Macroglobulinemia
- Author
-
Christopher J. Patterson, Jenny Sun, Guang Yang, Bryan Ciccarelli, Mariateresa Fulciniti, Owen A. O'Connor, Leukothea Ioakimidis, Xia Liu, Patricia Sheehy, Yangsheng Zhou, Lian Xu, Nikhil C. Munshi, Kaveh Maghsoudi, Hsuyi Tseng, Biao Zhu, Evdoxia Hatjiharissi, Ping Gong, Steven P. Treon, and Zachary R. Hunter
- Subjects
Male ,Cancer Research ,Cell Survival ,Immunoblotting ,Apoptosis ,Hydroxamic Acids ,Polymerase Chain Reaction ,Histone Deacetylases ,Bortezomib ,chemistry.chemical_compound ,Bone Marrow ,Cell Line, Tumor ,Panobinostat ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,business.industry ,Cell Cycle ,HDAC9 ,Macroglobulinemia ,Hematology ,Cell cycle ,Boronic Acids ,HDAC4 ,Histone Deacetylase Inhibitors ,Trichostatin A ,Oncology ,chemistry ,Pyrazines ,Cancer research ,Female ,Histone deacetylase ,Waldenstrom Macroglobulinemia ,business ,medicine.drug - Abstract
We studied the role of histone deacetylase inhibitors in Waldenstrom's macroglobulinemia (WM). Gene expression profiling of bone marrow CD19+ cells from 30 patients and 10 healthy donors showed overexpression of HDAC4, HDAC9, and Sirt5, with validation of HDAC9 overexpression by q-PCR in primary and BCWM.1 cells. Suberoylanilide hydroxamic acid, trichostatin A, panobinostat, and sirtinol demonstrated dose-dependent killing of BCWM.1 cells. TSA showed the greatest potency with IC50 of 70 nM. Importantly, HDAC9 activity was decreased following TSA treatment suggesting an essential role for this HDAC in WM therapy. The combination of bortezomib plus HDAC inhibitors resulted in at least additive tumor cell killing in BCWM.1 cells. TSA and bortezomib-induced apoptosis depended on a similar set of caspase activation, whereas their effect on cell cycle regulators was distinctly different. These results provided a framework for examining HDAC inhibitors as monotherapy, as well as combination therapy with bortezomib in WM.
- Published
- 2011
- Full Text
- View/download PDF
43. Second Primary Malignancies and Disease Transformation in Newly Diagnosed Symptomatic Patients with Waldenstrom's Macroglobulinemia: An Analysis from the Greek Myeloma Study Group
- Author
-
Nikolaos Giannakoulas, Dimitrios Christoulas, Panagiotis Tsirigotis, Michalis Michael, Elina Vervessou, Georgia Kaiafa, Eirini Katodritou, Evridiki Michali, Anastasia Pouli, Argiris Symeonidis, Maria Gavriatopoulou, Michail Iskas, Efstathios Kastritis, Amalia Vassou, Meletios A. Dimopoulos, Ioannis Ntanasis-Stathopoulos, Evdoxia Hatjiharissi, Panagiotis Repousis, Stavroula Giannouli, Emmanouil Spanoudakis, Evangelos Terpos, Marie-Christine Kyrtsonis, and Sossana Delibasi
- Subjects
medicine.medical_specialty ,business.industry ,Incidence (epidemiology) ,Immunology ,Waldenstrom macroglobulinemia ,Context (language use) ,Cell Biology ,Hematology ,Malignancy ,medicine.disease ,Biochemistry ,Interquartile range ,Median follow-up ,Internal medicine ,medicine ,Rituximab ,Cumulative incidence ,business ,medicine.drug - Abstract
Waldenström's Macroglobulinemia (WM) is an indolent lymphoma with heterogeneous clinical presentation and prolonged course. In this context, determining the overall disease and therapy burden in terms of complications and associated comorbidities is necessary. Of special interest is the occurrence of a second malignancy and transformation to high grade lymphoma, due to their impact on survival and quality of life but also on health resource use and follow up strategies. Several factors including immune dysfunction, genetic predisposition, environmental factors and treatment effects have been suggested as possible underlying pathophysiologic mechanisms for development of second primary malignancies or transformation in patients with WM. While there have been data published from various series, it is crucial to have data after prolonged follow up, as the time for development of such complications may be long. The current study included all patients with symptomatic WM that have been registered in the prospectively maintained database of the Greek Myeloma Study Group, in which the development of a second malignancy and disease transformation is being recorded, including type of second cancer and date of diagnosis. All patients fulfilled criteria for symptomatic WM diagnosis and for treatment initiation according to the Consensus Recommendations. The analysis included 598 symptomatic newly diagnosed patients with WM with a median follow up of 114 months. The median age was 69 years and 284 (47%) have died. Primary therapy was rituximab-based in 65%, contained alkylating agents in 78%, a nucleoside analogue in 2% and an anthracycline in 4%. In 46 patients (7.7%) the diagnosis of a second tumor was made after the initiation of treatment for symptomatic WM. The most common malignancies were cancer of the prostate (18%), stomach (6%), colon (6%), lung (6%), pancreas (3%), non-melanoma skin (3%) and central nervous system (6%) and AML/MDS (6%). In addition, in 12 (2%) patients, a diagnosis of another malignancy preceded the diagnosis of WM. The median time from treatment initiation for symptomatic WM to the diagnosis of a second primary malignancy was 51 months [interquartile range (IQR): 16-79]. The incidence ratio (IR) of a second primary malignancy was 0.0095 per person-years and the cumulative incidence of a second primary malignancy, accounting for death due to WM or other causes as a competing event, at 5 and 10 years was 3.6% and 6.6%, while, the risk of death from WM or other causes was 22.5% and 46%, respectively (Figure). The IR of a second malignancy was higher in men [IR ratio (IRR): 0.5, 95% CI 0.2-1.1, p=0.067] while it was similar for younger vs older patients (>75 years) (IRR: 1.57, 95% CI 0.61-3.5, p=0.27). Importantly, the use of rituximab as primary treatment was not associated with any increased risk of a second malignancy (IRR: 0.99, 95%CI 0.5-2.1, p=0.97). The incidence of second malignancies has not changed significantly in the era before vs after 2000. Overall, 19 (3.1%) transformation events were identified; the median time from treatment initiation to transformation was 51 months (IQR: 28-106). The IR for transformation was 0.005 per person-years and the cumulative risk for transformation, accounting for death of any cause as a competing event, was 2.2% and 3.6% at 5 and 10 years respectively. The presence of anemia (hemoglobin We conclude that the incidence of a second malignancy among patients with symptomatic WM is 7.7%, corresponding to an incidence rate of about 1 case per 100 patients per year. Transformation occurred in 3.1% corresponding to an incidence rate of 0.5 cases per 100 patients per year; this risk was higher among patients presenting with anemia and has been reduced after 2000. This may be related to the extensive use of rituximab and less frequent use of alkylating agents and nucleoside analogues. Figure. Figure. Disclosures Kastritis: Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Prothena: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Terpos:Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: member of steering committee, Research Funding; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel grant, Research Funding; BMS: Consultancy; Novartis: Consultancy; Genesis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel grant, Research Funding; Amgen Inc.: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel grant, steering committee member, Research Funding; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: member of DMC, Research Funding. Dimopoulos:Takeda: Honoraria; Celgene: Honoraria; Janssen: Honoraria; Bristol-Myers Squibb: Honoraria; Amgen: Honoraria.
- Published
- 2018
- Full Text
- View/download PDF
44. Expression of regulatory genes for lymphoplasmacytic cell differentiation in Waldenstrom Macroglobulinemia
- Author
-
Christopher J. Patterson, Steven P. Treon, Xavier Leleu, Irene M. Ghobrial, Evdoxia Hatjiharissi, Jacob D. Soumerai, Vinod Bakthavachalam, Zachary R. Hunter, Allen W. Ho, Sigitas Verselis, Edward A. Fox, Ruben D. Carrasco, Aldo M. Roccaro, Robert Manning, Anne-Sophie Moreau, Susanna Hamilton, Sophia Adamia, Daniel Ditzel Santos, and Lian Xu
- Subjects
Adult ,Male ,XBP1 ,Cellular differentiation ,Plasma Cells ,Gene Expression ,Biology ,Statistics, Nonparametric ,Genes, Regulator ,PRDM1 ,Gene expression ,medicine ,Humans ,Gene ,Aged ,Regulator gene ,Aged, 80 and over ,B-Lymphocytes ,Reverse Transcriptase Polymerase Chain Reaction ,Electrophoresis, Capillary ,Waldenstrom macroglobulinemia ,Cell Differentiation ,Sequence Analysis, DNA ,Hematology ,Middle Aged ,medicine.disease ,Reverse transcription polymerase chain reaction ,Case-Control Studies ,Cancer research ,Female ,Waldenstrom Macroglobulinemia - Abstract
Waldenstrom Macroglobulinemia (WM) is a B-cell malignancy characterized by excess bone marrow (BM) lymphoplasmacytic cells (LPC). The accumulation of LPC in WM may represent a failure of B-cells to properly differentiate into plasma cells. The present study investigated transcriptional expression of genes involved in late B-cell differentiation, including PRDM1, PAX5, XBP1 transcripts and ERN1, in BM B-cells from 31 patients with WM and six healthy donors. Real time reverse transcription polymerase chain reaction (RT-PCR) determined that approximately 80% of the patients had high XBP1 spliced mRNA expression, 80% of whom had high mRNA ERN1alpha expression. XBP1, PRDM1 and PAX5 mRNA was present in all patients studied. Using relative quantitative RT-PCR we isolated two groups with low and high expression of XBP1, XBP1 spliced and ERN1alpha. Sequence analysis showed germline polymorphisms in all genes studied. These data depict for the first time a heterogeneous expression pattern of the genes involved in late differentiation process of plasma cells in patients with WM and propose a role of XBP1-ERN1alpha in WM pathogenesis.
- Published
- 2009
- Full Text
- View/download PDF
45. Thalidomide and rituximab in Waldenstrom macroglobulinemia
- Author
-
Mark W. Pasmantier, Andrew R. Branagan, Henry Sonneborn, Bryan Ciccarelli, Jacob D. Soumerai, Zachary R. Hunter, David R. Lovett, Leukothea Ioakimidis, Kenneth C. Anderson, John M. Howard, Robert B. Cooper, Paul Musto, Hans Boedeker, Steven P. Treon, Alan Rauch, Cynthia Chua, Lawrence Garbo, Luis Chu, Maria Moore, John M. Hill, Frederick M. Briccetti, Stephen H. Nantel, Evdoxia Hatjiharissi, Harvey Zimbler, and Christopher J. Patterson
- Subjects
Adult ,Male ,medicine.medical_specialty ,Clinical Trials and Observations ,Immunology ,Phases of clinical research ,Hematocrit ,Biochemistry ,Gastroenterology ,Disease-Free Survival ,Drug Administration Schedule ,Antibodies, Monoclonal, Murine-Derived ,Internal medicine ,Immunopathology ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Adverse effect ,Aged ,Aged, 80 and over ,Dose-Response Relationship, Drug ,medicine.diagnostic_test ,business.industry ,Receptors, IgG ,Antibodies, Monoclonal ,Waldenstrom macroglobulinemia ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Neoadjuvant Therapy ,Thalidomide ,Surgery ,Treatment Outcome ,Peripheral neuropathy ,Immunoglobulin M ,Female ,Rituximab ,Waldenstrom Macroglobulinemia ,business ,Follow-Up Studies ,medicine.drug - Abstract
Thalidomide enhances rituximab-mediated, antibody-dependent, cell-mediated cytotoxicity. We therefore conducted a phase 2 study using thalidomide and rituximab in symptomatic Waldenstrom macroglobulinemia (WM) patients naive to either agent. Intended therapy consisted of daily thalidomide (200 mg for 2 weeks, then 400 mg for 50 weeks) and rituximab (375 mg/m2 per week) dosed on weeks 2 to 5 and 13 to 16. Twenty-five patients were enrolled, 20 of whom were untreated. Responses were complete response (n = 1), partial response (n = 15), and major response (n = 2), for overall and major response rate of 72% and 64%, respectively, on an intent-to-treat basis. Median serum IgM decreased from 3670 to 1590 mg/dL (P < .001), whereas median hematocrit rose from 33.0% to 37.6% (P = .004) at best response. Median time to progression for responders was 38 months. Peripheral neuropathy to thalidomide was the most common adverse event. Among 11 patients experiencing grade 2 or greater neuropathy, 10 resolved to grade 1 or less at a median of 6.7 months. Thalidomide in combination with rituximab is active and produces long-term responses in WM. Lower doses of thalidomide (ie, ≤ 200 mg/day) should be considered given the high frequency of treatment-related neuropathy in this patient population. This trial is registered at www.clinicaltrials.gov as #NCT00142116.
- Published
- 2008
- Full Text
- View/download PDF
46. The HMG-CoA inhibitor, simvastatin, triggersin vitroanti-tumour effect and decreases IgM secretion in Waldenstrom macroglobulinaemia
- Author
-
Aldo M. Roccaro, Christopher J. Patterson, Bryan Ciccarelli, Irene M. Ghobrial, Xiaoying Jia, Xavier Leleu, Hai T. Ngo, Steven P. Treon, Jacob D. Soumerai, Zachary R. Hunter, Robert Manning, Kelly O’Connor, Anne-Sophie Moreau, Lian Xu, Evdoxia Hatjiharissi, and Antonio Sacco
- Subjects
MAPK/ERK pathway ,Simvastatin ,medicine.medical_specialty ,Apoptosis ,In Vitro Techniques ,Biology ,CD19 ,Internal medicine ,medicine ,Humans ,Extracellular Signal-Regulated MAP Kinases ,Protein kinase A ,Protein kinase B ,Kinase ,Cell growth ,Hematology ,Endocrinology ,Immunoglobulin M ,HMG-CoA reductase ,biology.protein ,Cancer research ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Waldenstrom Macroglobulinemia ,Proto-Oncogene Proteins c-akt ,medicine.drug - Abstract
Summary Waldenstrom macroglobulinaemia (WM) is an incurable lymphoplasmacytic lymphoma with secretion of serum monoclonal immunoglobulin M (IgM). We previously showed that patients receiving cholesterol-lowering statins, had the lowest IgM value in a large cohort of patients with WM. Simvastatin, a 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitor, induced inhibition of proliferation, cytotoxic effect and apoptosis in IgM secreting cell lines as well as in primary CD19 + WM cells. Interestingly, those effects were reversed by addition of mevalonate and geranylgeranylpyrophosphate, demonstrating that simvastatin inhibited cell growth, survival and IgM secretion on BCWM.1 WM cells by inhibition of geranylgeranylated proteins. Furthermore, simvastatin overcame tumour cell growth induced by co-culture of WM cells with bone-marrow stromal cells. Simvastatin also decreased IgM secretion by BCWM.1 cells at an early time-point that had not affected cell survival. Simvastatin-induced cytotoxicity was preceded by a decrease in Akt (protein kinase B, PKB) and extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) pathways at 18 h. In addition, simvastatin induced an increase in stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/ JNK) MAPK followed by caspase-8, -9, -3 and poly(ADP-ribose) polymerase (PARP) cleavages at 18 h, leading to apoptosis. Furthermore, simvastatin enhanced the cytotoxicity induced by bortezomib, fludarabine and dexamethasone. Our studies therefore support our earlier observation of statin-mediated anti-WM activity and provide the framework for future clinical trials testing simvastatin in WM.
- Published
- 2008
- Full Text
- View/download PDF
47. Targeting Akt and Heat Shock Protein 90 Produces Synergistic Multiple Myeloma Cell Cytotoxicity in the Bone Marrow Microenvironment
- Author
-
Judith Runnels, Kenneth C. Anderson, Yazan Alsayed, Teru Hideshima, Irene M. Ghobrial, Aldo M. Roccaro, Alissa Huston, David Roodman, Yu Tsu Tai, Nikhil C. Munshi, Peter Sportelli, Xavier Leleu, Judy Anderson, Paul G. Richardson, Anne Moreau, Xiaoying Jia, Evdoxia Hatjiharissi, Sonia Vallet, and Hai T. Ngo
- Subjects
DNA Replication ,Cancer Research ,Stromal cell ,Cell Survival ,Angiogenesis ,Lactams, Macrocyclic ,Phosphorylcholine ,Biology ,chemistry.chemical_compound ,Bone Marrow ,Osteoclast ,Cell Line, Tumor ,Benzoquinones ,medicine ,Humans ,HSP90 Heat-Shock Proteins ,Protein kinase B ,Cell Cycle ,Flow Cytometry ,Perifosine ,Vascular endothelial growth factor ,medicine.anatomical_structure ,Oncology ,chemistry ,Cancer research ,Bone marrow ,Growth inhibition ,Multiple Myeloma ,Proto-Oncogene Proteins c-akt ,Cell Division - Abstract
Purpose: We hypothesized that targeting both Akt and heat shock protein (HSP) 90 would induce cytotoxic activity against multiple myeloma (MM) cells and target the bone marrow (BM) microenvironment to inhibit angiogenesis, osteoclast formation, as well as migration and adhesion of MM cells. Experimental Design: MM cell lines were incubated with perifosine (5 and 10 μmol/L) and 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (17-DMAG; 50 and 100 nmol/L) alone and in combination. Results: The combination of Akt inhibitor perifosine and HSP90 inhibitor 17-DMAG was synergistic in inducing MM cell cytotoxicity, evidenced by inhibition of DNA synthesis and induction of apoptosis. In addition, perifosine and 17-DMAG almost completely inhibited osteoclast formation: perifosine interfered with both early and late stages of osteoclast progenitor development, whereas 17-DMAG targeted only early stages. We next showed that combined therapy overcomes tumor growth and resistance induced by BM stromal cells and endothelial cells as well as the proliferative effect of exogenous interleukin-6, insulin-like growth factor-I, and vascular endothelial growth factor. Moreover, the combination also induced apoptosis and growth inhibition in endothelial cells and inhibited angiogenesis. Finally, we showed that the two agents prevented migration of MM cells toward stromal-derived factor-1 and vascular endothelial growth factor, which are present in the BM milieu, and also prevented adhesion of MM cells to fibronectin. Conclusions: This study provides the preclinical framework for treatment protocols targeting both the Akt and HSP pathways in MM.
- Published
- 2008
- Full Text
- View/download PDF
48. Genetic Linkage of FcγRIIa and FcγRIIIa and Implications for Their Use in Predicting Clinical Responses to CD20-Directed Monoclonal Antibody Therapy
- Author
-
Edward A. Fox, Steven P. Treon, Alexandros Kortsaris, Mark Hansen, Daniel Ditzel Santos, Christopher J. Patterson, Sigitas Verselis, Zachary R. Hunter, Evdoxia Hatjiharissi, Xavier Leleu, Elizabeth W. Dimmock, Allen W. Ho, Andrew R. Branagan, and Lian Xu
- Subjects
CD20 ,biology ,business.industry ,Genetic linkage ,Immunology ,biology.protein ,Medicine ,General Medicine ,business ,Monoclonal antibody therapy - Published
- 2007
- Full Text
- View/download PDF
49. Novel Agents in the Treatment of Waldenström's Macroglobulinemia
- Author
-
Christopher J. Patterson, Robert Manning, Evdoxia Hatjiharissi, Cao Yang, Jacob D. Soumerai, Zachary R. Hunter, Antonio Sacco, Lian Xu, Bryan Ciccarelli, Allen W. Ho, Andrew R. Branagan, Olivier Tournilhac, Irene M. Ghobrial, Hai T. Ngo, Kelly O’Connor, Xavier Leleu, Steven P. Treon, Aldo M. Roccaro, Sophia Adamia, Daniel Ditzel Santos, Xiaoying Jia, Marybeth Nelson, Renee Leduc, and Anne-Sophie Moreau
- Subjects
Cancer Research ,CD52 ,Antineoplastic Agents ,Autologous stem-cell transplantation ,immune system diseases ,hemic and lymphatic diseases ,Gammopathy ,medicine ,Animals ,Humans ,Immunologic Factors ,Protease Inhibitors ,cardiovascular diseases ,Lenalidomide ,Bortezomib ,business.industry ,Macroglobulinemia ,Hematology ,General Medicine ,Oncology ,Immunology ,Alemtuzumab ,Rituximab ,Waldenstrom Macroglobulinemia ,business ,medicine.drug - Abstract
Waldenström's macroglobulinemia is a B-cell disorder characterized by bone marrow infiltration with lymphoplasmacytic cells and demonstration of an immunoglobulin M monoclonal gammopathy. Despite advances in therapy, Waldenström's macroglobulinemia remains incurable. As such, novel therapeutic agents are needed for the treatment of Waldenström's macroglobulinemia. In ongoing efforts, we and others have sought to exploit advances made in the understanding of the biology of Waldenström's macroglobulinemia so as to better target therapeutics for this malignancy. Importantly, as part of these efforts, we have prioritized the development of stem cell-sparing drugs because autologous stem cell transplantation remains a viable salvage option in Waldenström's macroglobulinemia. These efforts have led to the development of several novel agents for treating Waldenström's macroglobulinemia, including bortezomib; monoclonal antibodies and/or blocking protein targeting CD40, CD52, or CD70, a proliferation-inducing ligand and B-lymphocyte stimulator; the immunomodulator thalidomide as an enhancer of rituximab activity, as well as agents interfering with stem cell factor, phosphatidylinositol 3-kinase/Akt, phosphodiesterase, cholesterol, and protein kinase C beta signaling. This report provides an update on biologic studies and clinical efforts for the development of these novel agents in the treatment of Waldenström's macroglobulinemia.
- Published
- 2007
- Full Text
- View/download PDF
50. Competing risk survival analysis in patients with symptomatic Waldenström macroglobulinemia: the impact of disease unrelated mortality and of rituximab-based primary therapy
- Author
-
Anastasia Sioni, Elina Vervessou, Efstathios Kastritis, Michail Michael, Maria Gavriatopoulou, Meletios A. Dimopoulos, Panagiotis Repousis, Constantinos Tsatalas, Pierre Morel, Amalia Vassou, Argirios S. Symeonidis, Evdoxia Hatjiharissi, Marie-Christine Kyrtsonis, Evrydiki Michalis, Evangelos Terpos, Sossana Delimpasi, and Michael Voulgarelis
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,Cyclophosphamide ,Disease ,Models, Biological ,Disease-Free Survival ,Risk Factors ,medicine ,Humans ,Online Only Articles ,Survival rate ,Survival analysis ,Aged ,Aged, 80 and over ,business.industry ,Waldenstrom macroglobulinemia ,Hematology ,Middle Aged ,medicine.disease ,Lymphoma ,Fludarabine ,Surgery ,Survival Rate ,Rituximab ,Female ,Waldenstrom Macroglobulinemia ,business ,medicine.drug ,Follow-Up Studies - Abstract
WM is a disease of the elderly with a protracted course in many patients and a median survival of 7 to 10 years,[1][1] however, in many patients, other factors rather than WM or its treatment may be the cause of death.[2][2] Non-WM-related mortality mainly affects outcomes of elderly patients and is
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.