99 results on '"S. Manier"'
Search Results
2. S176: DARATUMUMAB CARFILZOMIB LENALIDOMIDE AND DEXAMETHASONE AS INDUCTION THERAPY IN HIGH-RISK TRANSPLANT ELIGIBLE NEWLY DIAGNOSED MYELOMA PATIENTS: RESULTS OF THE PHASE 2 STUDY IFM 2018-04
- Author
-
C. Touzeau, A. Perrrot, C. Hulin, S. Manier, M. Macro, M.-L. Chretien, L. Karlin, O. Decaux, C. Jacquet, M. Tiab, X. Leleu, L. Planchce, H. Avet-Loiseau, and P. Moreau
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
3. S180: RG6234, A NOVEL GPRC5D T-CELL ENGAGING BISPECIFIC ANTIBODY, INDUCES RAPID RESPONSES IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA: PRELIMINARY RESULTS FROM A FIRST-IN-HUMAN TRIAL
- Author
-
C. Hasselbalch Riley, M. Hutchings, S.-S. Yoon, Y. Koh, S. Manier, T. Facon, S. J. Harrison, J. Er, F. Volzone, A. Pinto, C. Montes, E. M. Ocio, A. Alfonso-Pierola, P. Rodriguez Otero, F. Offner, A. Guidetti, P. Corradini, C. Titouan, C. Hulin, C. Touzeau, P. Moreau, R. Popat, S. Leong, R. Mazza, A.-M. E. Bröske, I. Dekhtiarenko, H.-J. Helms, S. Belli, T. Vardar, T. Fauti, J. Eckmann, T. Moore, M. Schneider, W. Jacob, M. Weisser, and C. Carlo-Stella
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
4. S184: EVALUATING TECLISTAMAB IN PATIENTS WITH RELAPSED/ REFRACTORY MULTIPLE MYELOMA FOLLOWING EXPOSURE TO OTHER B-CELL MATURATION ANTIGEN (BCMA)-TARGETED AGENTS
- Author
-
C. Touzeau, A. Krishnan, P. Moreau, A. Perrot, S. Z. Usmani, S. Manier, M. Cavo, C. Martinez-Chamorro, A. Nooka, T. Martin, L. Karlin, X. Leleu, N. Bahlis, B. Besemer, L. Pei, R. Verona, S. Girgis, C. Uhlar, R. Kobos, and A. Garfall
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
5. S173: T-CELL ACTIVATION AND MYELOMA CELL KILLING CONFIRM THE MODE OF ACTION OF RG6234, A NOVEL GPRC5D T-CELL ENGAGING BISPECIFIC ANTIBODY, IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA
- Author
-
I. Dekhtiarenko, I. Lelios, J. Attig, N. Sleiman, D. Lazzaro, I. Clausen, N. Gräfe, H.-J. Helms, E. Schindler, S. Belli, T. Fauti, J. Eckmann, P. Umana, W. Jacob, M. Schneider, C. Hasselbalch Riley, M. Hutchings, S.-S. Yoon, Y Koh, S. Manier, T. Facon, S. J. Harrison, J. Er, F. Volzone, A. Pinto, C. Montes, E. M. Ocio, A. Alfonso-Pierola, P. Rodríguez Otero, F. Offner, A. Guidetti, P. Corradini, C. Titouan, C. Hulin, C. Touzeau, P. Moreau, R. Popat, S. Leong, R. Mazza, C. Carlo-Stella, and A.-M. E. Bröske
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
6. P891: IXAZOMIB, POMALIDOMIDE AND DEXAMETHASONE (IXPD) IN RELAPSED OR REFRACTORY MULTIPLE MYELOMA (RRMM) CHARACTERIZED WITH HIGH-RISK CYTOGENETICS. IFM 2014-01
- Author
-
A. Bobin, S. Manier, J. De Keizer, L. Karlin, A. Perrot, C. Hulin, D. Caillot, C. Mariette, P. Lenain, V. Richez, M. Tiab, C. Touzeau, A. Jaccard, O. Decaux, C. Araujo, K. Belhadj, L. Benboubker, C. Déal, M. Macro, L. Vincent, B. Arnulf, B. Bareau, T. Braun, C. Calmettes, D. mamoun, H. Zerazhi, H. Demarquette, P. Feugier, C. Fohrer-sonntag, S. Godet, M.-O. Petillon, H. Avet-Loiseau, and X. Leleu
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
7. P851: TARGETING GENE DEPENDENCIES IN MYC OVEREXPRESSING MULTIPLE MYELOMA
- Author
-
L. Hasan Bou Issa, L. Fléchon, W. Laine, A. Ouelkdite, G. Escure, S. Gaggero, T. Ingegnere, R. Sklavenitis Pistofidis, I. M. Ghobrial, T. Facon, S. Mitra, J. Kluza, B. Quesnel, and S. Manier
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
8. P872: DEL(1P32) REMAINS A POWERFUL PROGNOSTIC FACTOR IN A LARGE COHORT OF NDMM PATIENTS: AN UPDATE
- Author
-
A. Schavgoulidze, A. Perrot, T. Cazaubiel, X. Leleu, S. Manier, L. Buisson, S. Maheo, L. Do Souto Ferreira, R. Lannes, L. Pavageau, C. Hulin, J.-P. Marolleau, L. Voillat, K. Belhadj, M. Divoux, B. Slama, S. Brechignac, M. Macro, A.-M. Stoppa, L. Sanhes, F. Orsini-Piocelle, J. Fontan, M.-L. Chretien, H. Demarquette, M. Mohty, H. Avet-Loiseau, and J. Corre
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
9. P866: CORRELATIVE ANALYSIS TO DEFINE PATIENT PROFILES ASSOCIATED WITH MANUFACTURING AND CLINICAL ENDPOINTS IN RELAPSED/REFRACTORY MULTIPLE MYELOMA PATIENTS TREATED WITH IDECABTAGENE VICLEUCEL (BB2121)
- Author
-
J. Rytlewski, J. Fuller, D. R. Mertz, C. Freeman, S. Manier, N. Shah, and T. B. Campbell
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
10. P929: IXAZOMIB AND DARATUMUMAB WITHOUT DEXAMETHASONE (I-DARA) IN ELDERLY FRAIL RELAPSING MYELOMA (RRMM) PATIENTS: RESULTS OF THE PHASE 2 STUDY IFM 2018-02 OF THE INTERGROUPE FRANCOPHONE DU MYELOME (IFM).
- Author
-
M. Macro, C. Touzeau, C. Mariette, S. Manier, S. Brechignac, L. Vincent, B. Hebraud, O. Decaux, S. Schulmann, C. Lenoir, P. Godmer, A. Farge, L. Peyro Saint Paul, J.-J. Parienti, and X. Leleu
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
11. P957: CARFILZOMIB, LENALIDOMIDE, DEXAMETHASONE FOLLOWED BY A SECOND AUTO-HCT IS AN EFFECTIVE STRATEGY IN FIRST RELAPSE MULTIPLE MYELOMA: A STUDY OF THE CHRONIC MALIGNANCIES WORKING PARTY OF EBMT
- Author
-
R. Tilmont, I. Yakoub-Agha, D.-J. Eikema, N. Zinger, M. Haenel, N. Schaap, C. Herrera Arroyo, C. Schuermans, W. Bethge, M. Engelhardt, J. Kuball, M. Michieli, N. Schub, K. M. O. Wilson, J. H. Bourhis, M. V. Mateos, N. Robin, E. Jost, N. Kröger, J. M. Moraleda, S. Sica, P. J. Hayden, M. Beksac, S. Schönland, and S. Manier
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
12. P1257: MOLECULAR CHARACTERIZATION AND CLONAL EVOLUTION OF MYCOSIS FUNGOIDES
- Author
-
L. Fléchon, I. Arib, A. Dutta, R. Sklanvenitis Pistofidis, C. Stewart, R. Dubois, S. Poulain, M.-C. Copin, S. Javed, M. Nudel, L. Hasan Bou Issa, A. Ouelkite-Oumouchal, S. Faiz, O. Carpentier, L. Mortier, S. Mitra, M. Figeac, F. Morschhauser, B. Quesnel, I. Ghobrial, and S. Manier
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2022
- Full Text
- View/download PDF
13. Whole-exome sequencing of cell-free DNA and circulating tumor cells in multiple myeloma
- Author
-
S. Manier, J. Park, M. Capelletti, M. Bustoros, S. S. Freeman, G. Ha, J. Rhoades, C. J. Liu, D. Huynh, S. C. Reed, G. Gydush, K. Z. Salem, D. Rotem, C. Freymond, A. Yosef, A. Perilla-Glen, L. Garderet, E. M. Van Allen, S. Kumar, J. C. Love, G. Getz, V. A. Adalsteinsson, and I. M. Ghobrial
- Subjects
Science - Abstract
Circulating tumor cells (CTCs) and cell-free DNA (cfDNA) enables characterization of a patient’s cancer. Here, the authors analyse CTCs, cfDNA, and tumor biopsies from multiple myeloma patients to show these approaches are complementary for mutation detection, together enabling a greater fraction of patient tumors to be profiled.
- Published
- 2018
- Full Text
- View/download PDF
14. Myélome multiple du sujet âgé : Entre évaluation de la fragilité des patients et progrès de l’Immunothérapie
- Author
-
T. Facon and S. Manier
- Subjects
General Medicine - Published
- 2022
15. Longitudinal study of AMH variations in 122 Adolescents and Young Adults (AYA) and non-AYA lymphoma patients to evaluate the chemo-induced ovarian toxicity to further personalise fertility preservation counselling
- Author
-
C Robin, Franck Morschhauser, M Barbatti, Pascal Pigny, Christine Decanter, J Labreuche, B Bruno, S Manier, H Behal, and J Delepine
- Subjects
Oncology ,Adult ,Anti-Mullerian Hormone ,Counseling ,medicine.medical_specialty ,Adolescent ,Vinblastine ,Bleomycin ,Young Adult ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Fertility preservation ,Longitudinal Studies ,Prospective Studies ,Young adult ,Prospective cohort study ,business.industry ,Rehabilitation ,Obstetrics and Gynecology ,Repeated measures design ,Fertility Preservation ,Chemotherapy regimen ,Hodgkin Disease ,Dacarbazine ,Regimen ,Reproductive Medicine ,ABVD ,Doxorubicin ,Population study ,Female ,business ,medicine.drug - Abstract
STUDY QUESTION What is the influence of age and chemotherapy regimen on the longitudinal blood anti-Müllerian hormone (AMH) variations in a large series of adolescents and young adult (AYA) (15–24 years old) and non-AYA (25–35 years old) lymphoma patients? SUMMARY ANSWER In case of alkylating regimen treatment, there was a deep and sustained follicular depletion in AYA as well as non-AYA patients; however in both groups, the ovarian toxicity was extremely low in cases of non-alkylating treatments. WHAT IS KNOWN ALREADY AMH is now well-recognised to be a real-time indicator of ovarian follicular depletion and recovery in women treated by chemotherapy. Its longitudinal variations may discriminate between highly and minimally toxic protocols regarding ovarian function. It has been shown, in different cancer types, that age, type of chemotherapy regimen and pre-treatment AMH levels are the main predictors of ovarian recovery. Large studies on longitudinal AMH variations under chemotherapy in lymphoma patients are few but can provide the opportunity to assess the degree of follicle loss at a young age. STUDY DESIGN, SIZE, DURATION This prospective cohort study was conducted in the Fertility Observatory of the Lille University Hospital. Data were collected between 2007 and 2016. Non-Hodgkin or Hodgkin lymphoma patients (n = 122) between 15 and 35 years old were prospectively recruited before commencing chemotherapy. Patients were treated either by a non-alkylating protocol (ABVD group; n = 67) or by an alkylating regimen (alkylating group; n = 55). PARTICIPANTS/MATERIALS, SETTING, METHODS Serial AMH measurements were performed at baseline (AMH0), 15 days after the start of chemotherapy (AMH1), 15 days before the last chemotherapy cycle (AMH2), and at time 3, 6, 9, 12, 18 and 24 months from the end of chemotherapy. The whole study population was divided into two groups according to age: AYA (15–24; n = 65) and non-AYA (25–35; n = 57). All patients received a once monthly GnRH agonist injection during the whole treatment period. A linear mixed model was used to account for the repeated measures of single patients. MAIN RESULTS AND THE ROLE OF CHANCE At baseline, non-AYA patients had higher BMI and lower AMH levels than AYA patients. All AYA and non-AYA patients having received ABVD protocols had regular cycles at 12 months of follow-up. In case of alkylating regimens, amenorrhoea was more frequent in non-AYA patients than in AYA patients at 12 months (37% vs 4%, P = 0.011) and at 24 months (24% vs 4%, P = 0.045). We distinguished a similar depletion phase from AMH0 to AMH2 between ABVD and alkylating groups but significantly different recovery phases from AMH2 to AMH + 24 months. AMH recovery was fast and complete in case of ABVD protocols whatever the age: AMH reached pre-treatment values as soon as the 6th month of follow-up in the AYA group (mean (95% CI) in log AMH M0 vs M6: 3.07 (2.86 to 3.27) vs 3.05 (2.78 to 3.31), P = 1.00) and in the non-AYA group (mean (95% CI) in log AMH M0 vs M6: 2.73 (2.40 to 3.05) vs 2.47 (2.21 to 2.74), P = 1.00). In contrast, no patients from the alkylating group returned to pre-treatment AMH values whatever the age of patients (AYA or non-AYA). Moreover, none of the AMH values post-chemotherapy in the non-AYA group were significantly different from AMH2. Conversely in the AYA group, AMH levels from 6 months (mean (95% CI) in log AMH: 1.79 (1.47 to 2.11), P LIMITATIONS, REASONS FOR CAUTION There was a large disparity in subtypes of protocols in the alkylating group. The average duration of chemotherapy for patients treated with alkylating protocols was longer than that for patients treated with ABVD. WIDER IMPLICATIONS OF THE FINDINGS These results make it possible to develop strategies for fertility preservation according to age and type of protocol in a large series of young lymphoma patients. In addition, it was confirmed that young age does not protect against ovarian damage caused by alkylating agents. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by Agence Régionale de Santé Hauts de France and Agence Onco Hauts-de-France who provided finances for AMH dosages (n° DOS/SDES/AR/FIR/2019/282). There are no competing interests. TRIAL REGISTRATION NUMBER DC-2008-642 and CNIL DEC2015-112.
- Published
- 2021
16. P13: CAN PATIENT-REPORTED OCULAR SYMPTOMS GUIDE DOSE MODIFICATIONS IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA RECEIVING BELANTAMAB MAFODOTIN?
- Author
-
R Popat, A Badros, S Kumar, P Rodriguez-Otero, A Cohen, S Manier, P Voorhees, F Gay, R Rifkin, T Martin, A Chari, K Weisel, A Farooq, B Jeng, W Chng, H Lee, J Berdeja, V Jadhav, A Tosolini, L Eliason, A Palumbo, M Dimopoulos, S Lonial, S Trudel, P Richardson, and E Terpos
- Subjects
Hematology - Published
- 2022
17. S1600 REAL-WORLD RESULTS ON CD19 CAR T-CELL FOR 60 FRENCH PATIENTS WITH RELAPSED/REFRACTORY DIFFUSE LARGE B-CELL LYMPHOMA INCLUDED IN A TEMPORARY AUTHORIZATION FOR USE (ATU) PROGRAM
- Author
-
Gilles Salles, R. Di Blasi, Sophie Bernard, T. Kanounni, Steven Legouill, Catherine Thieblemont, Jérôme Paillassa, S. Manier, Nathalie Fegueux, Corinne Haioun, Guillaume Cartron, Pierre Sesques, Roch Houot, H. Tilly, F. Morschhauser, T. Gastine, Benoit Tessoulin, and Emmanuel Bachy
- Subjects
Oncology ,medicine.medical_specialty ,biology ,business.industry ,Authorization ,Hematology ,medicine.disease ,CD19 ,Internal medicine ,Relapsed refractory ,biology.protein ,medicine ,Car t cells ,business ,Diffuse large B-cell lymphoma - Published
- 2019
18. Targeting MYC in multiple myeloma
- Author
-
K K Jovanović, C Roche-Lestienne, I M Ghobrial, T Facon, B Quesnel, and S Manier
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2017
19. PF637 EFFICACY OF LENALIDOMIDE AND LOW-DOSE DEXAMETHASONE IN NEWLY DIAGNOSED PATIENTS WITH MULTIPLE MYELOMA: RESULTS FROM A META-ANALYSIS OF RANDOMIZED CONTROLLED TRIAL EVIDENCE
- Author
-
S. Robinson, T. Facon, S. Manier, G. Jackson, A. Frederickson, M. Owen, and S. Dhanasiri
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Low dose ,Hematology ,Newly diagnosed ,medicine.disease ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,Meta-analysis ,medicine ,business ,Dexamethasone ,Multiple myeloma ,medicine.drug ,Lenalidomide - Published
- 2019
20. EFFICACY AND SAFETY OF ELRANATAMAB IN PATIENTS WITH RELAPSED OR REFRACTORY MULTIPLE MYELOMA AND PRIOR B-CELL MATURATION ANTIGEN–DIRECTED THERAPIES: A POOLED ANALYSIS FROM MAGNETISMM STUDIES
- Author
-
A Nooka, A Lesokhin, M Mohty, R Niesvizky, C Maisel, B Arnulf, S Larson, A Varshavsky-Yanovsky, X Leleu, L Karlin, D Vesole, N Bahlis, CF Larrea, N Raje, E Leip, U Conte, M Elmeliegy, A Viqueira, V Blunk, and S Manier
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Introduction/Objectives: To evaluate the efficacy and safety of elranatamab in a pooled analysis of patients (pts) enrolled in MagnetisMM trials with relapsed or refractory multiple myeloma (RRMM) who had prior exposure to B-cell maturation antigen (BCMA)-directed therapy. Materials and methods: Eligible pts received at least 1 proteasome inhibitor, 1 immunomodulatory drug, 1 anti-CD38 antibody, and 1 BCMA-directed therapy (antibody-drug conjugate [ADC] and/or chimeric antigen receptor [CAR]-T cells). The pooled analysis included pts in the MagnetisMM-1 trial (NCT03269136; n = 13) who received subcutaneous (SC) elranatamab 215-1000 μg/kg; MM-3 (NCT04649359; n = 64) and MM-9 (NCT05014412; n = 9) who received the recommended phase 2 dose of 76 mg SC once-weekly. Efficacy endpoints were evaluated by investigator per IMWG criteria. TEAEs were graded by CTCAE (MM-1, v4.03; MM-3 & MM-9, v5.0); CRS and ICANS were graded by ASTCT criteria. Results include data up through ≍10 months after last pt initial dose in all pooled studies. Results: In total, 86 pts were included. Median age was 66.0 y (range, 40-84); 47.7% male. At baseline, 69.8% had an ECOG PS ≥1; 24.4% had high risk cytogenetics; 54.7% had extramedullary disease. Pts received a median of 7.0 (3-19) prior lines of therapy, including BCMA-directed ADC (67.4%), CAR T-cells (41.9%); 9.3% received both. 96.5% and 54.7% of pts were triple-class and penta-drug refractory, respectively; among pts who received ADC and CAR-T cells respectively, 79.3% and 27.8% were refractory to ADC and CAR-T cells. After a median follow-up of 10.3 mo (0.3-32.3), median duration of treatment was 3.3 mo (0.03-30.4). At the cut-off date, 24.4% of pts remained on treatment; most common reason for permanent treatment discontinuation was progressive disease (44.2%). The overall response rate (ORR) was 45.3% (95% CI 34.6-56.5), with ≥CR achieved in 17.4% of pts. ORR for pts with prior BCMA-directed ADC and CAR-T cells was 41.4% (95% CI 28.6-55.1) and 52.8% (95% CI 35.5-69.6), respectively. Among responders, median time to objective response was 1.9 mo (0.3-9.3). Median duration of response (DOR) was not reached by 10 mo; the DOR rate at 9 mo was 72.4% (95% CI 54.7-84.2). DOR rate (95% CI) for pts with prior BCMA-directed ADC and CAR-T cells were 67.3% (43.1-83.0) and 78.9% (53.2-91.5) at 9 mo, respectively. Median progression-free survival was 4.8 mo (95% CI 1.9-7.7); median overall survival was not reached by 10 mo, with a rate of 60.1% (95% CI 48.9-69.6) at 9 mo. Most common (≥25% of pts) TEAEs were CRS (65.1% [G3 1.2%]), anemia (59.3% [G3/4, 46.5%]), neutropenia (44.2% [G3/4, 40.7%]), thrombocytopenia (40.7% [G3/4, 29.1%]), diarrhea (33.7% [G3/4, 0%], and lymphopenia (32.6% [G3/4, 30.2%]). 5.8% (G3, 2.3%) of pts. Discussion: In pts with RRMM and prior exposure to BCMA-directed therapies, elranatamab was efficacious and well tolerated; no new safety signals were observed vs the BCMA-naïve population. Conclusions: These results support treatment with elranatamab in pts with RRMM post BCMA-directed therapy.
- Published
- 2023
- Full Text
- View/download PDF
21. EFFICACY AND SAFETY OF ELRANATAMAB BY AGE AND FRAILTY IN PATIENTS WITH RELAPSED/REFRACTORY MULTIPLE MYELOMA: A SUBGROUP ANALYSIS FROM MAGNETISMM-3
- Author
-
N Raje, X Leleu, A Lesokhin, M Mohty, A Nooka, E Leip, U Conte, A Viqueira, V Blunk, and S Manier
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Introduction/Objectives: Multiple myeloma is a disease of elderly and frail people, who are often predominantly ineligible for intensive therapies. The objective of this analysis is to report the efficacy and safety of elranatamab monotherapy by age and frailty in B-cell maturation antigen-naïve patients (pts) with relapsed or refractory multiple myeloma (RRMM) enrolled into cohort A of the ongoing phase 2 MagnetisMM -3 (NCT04649359) study. Materials and methods: Eligibility criteria, dosing and administration were previously reported (Bahlis et al, ASH 2022). Subgroups of pts within Cohort A (n = 123) were analyzed by age:
- Published
- 2023
- Full Text
- View/download PDF
22. Deciphering genetic and nongenetic factors underlying tumour dormancy: insights from multiomics analysis of two syngeneic MRD models of melanoma and leukemia.
- Author
-
Laguillaumie MO, Titah S, Guillemette A, Neve B, Leprêtre F, Ségard P, Shaik FA, Collard D, Gerbedoen JC, Fléchon L, Hasan Bou Issa L, Vincent A, Figeac M, Sebda S, Villenet C, Kluza J, Laine W, Fournier I, Gimeno JP, Wisztorski M, Manier S, Tarhan MC, Quesnel B, Idziorek T, and Touil Y
- Subjects
- Animals, Mice, Leukemia genetics, Leukemia pathology, DNA Copy Number Variations, Exome Sequencing, Mice, Inbred C57BL, Proteomics, Transcriptome, Gene Expression Profiling, Multiomics, Melanoma genetics, Melanoma pathology, Neoplasm, Residual, Disease Models, Animal
- Abstract
Background: Tumour dormancy, a resistance mechanism employed by cancer cells, is a significant challenge in cancer treatment, contributing to minimal residual disease (MRD) and potential relapse. Despite its clinical importance, the mechanisms underlying tumour dormancy and MRD remain unclear. In this study, we employed two syngeneic murine models of myeloid leukemia and melanoma to investigate the genetic, epigenetic, transcriptomic and protein signatures associated with tumour dormancy. We used a multiomics approach to elucidate the molecular mechanisms driving MRD and identify potential therapeutic targets., Results: We conducted an in-depth omics analysis encompassing whole-exome sequencing (WES), copy number variation (CNV) analysis, chromatin immunoprecipitation followed by sequencing (ChIP-seq), transcriptome and proteome investigations. WES analysis revealed a modest overlap of gene mutations between melanoma and leukemia dormancy models, with a significant number of mutated genes found exclusively in dormant cells. These exclusive genetic signatures suggest selective pressure during MRD, potentially conferring resistance to the microenvironment or therapies. CNV, histone marks and transcriptomic gene expression signatures combined with Gene Ontology (GO) enrichment analysis highlighted the potential functional roles of the mutated genes, providing insights into the pathways associated with MRD. In addition, we compared "murine MRD genes" profiles to the corresponding human disease through public datasets and highlighted common features according to disease progression. Proteomic analysis combined with multi-omics genetic investigations, revealed a dysregulated proteins signature in dormant cells with minimal genetic mechanism involvement. Pathway enrichment analysis revealed the metabolic, differentiation and cytoskeletal remodeling processes involved in MRD. Finally, we identified 11 common proteins differentially expressed in dormant cells from both pathologies., Conclusions: Our study underscores the complexity of tumour dormancy, implicating both genetic and nongenetic factors. By comparing genomic, transcriptomic, proteomic, and epigenomic datasets, our study provides a comprehensive understanding of the molecular landscape of minimal residual disease. These results provide a robust foundation for forthcoming investigations and offer potential avenues for the advancement of targeted MRD therapies in leukemia and melanoma patients, emphasizing the importance of considering both genetic and nongenetic factors in treatment strategies., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
23. Isatuximab, lenalidomide, dexamethasone and bortezomib in transplant-ineligible multiple myeloma: the randomized phase 3 BENEFIT trial.
- Author
-
Leleu X, Hulin C, Lambert J, Bobin A, Perrot A, Karlin L, Roussel M, Montes L, Cherel B, Chalopin T, Slama B, Chretien ML, Laribi K, Dingremont C, Roul C, Mariette C, Rigaudeau S, Calmettes C, Dib M, Tiab M, Vincent L, Delaunay J, Santagostino A, Macro M, Bourgeois E, Orsini-Piocelle F, Gay J, Bareau B, Bigot N, Vergez F, Lebreton P, Tabrizi R, Waultier-Rascalou A, Frenzel L, Le Calloch R, Chalayer E, Braun T, Lachenal F, Corm S, Kennel C, Belkhir R, Bladé JS, Joly B, Richez-Olivier V, Gardeney H, Demarquette H, Robu-Cretu D, Garderet L, Newinger-Porte M, Kasmi A, Royer B, Decaux O, Arnulf B, Belhadj K, Touzeau C, Mohty M, Manier S, Moreau P, Avet-Loiseau H, Corre J, and Facon T
- Subjects
- Humans, Aged, Male, Female, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized adverse effects, Neoplasm, Residual, Treatment Outcome, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Bortezomib administration & dosage, Bortezomib therapeutic use, Lenalidomide administration & dosage, Lenalidomide therapeutic use, Dexamethasone administration & dosage, Dexamethasone therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
CD38-targeting immunotherapy is approved in combination with lenalidomide and dexamethasone in patients with newly diagnosed multiple myeloma (NDMM) that are transplant ineligible (TI) and is considered the best standard of care (SOC). To improve current SOC, we evaluated the added value of weekly bortezomib (V) to isatuximab plus lenalidomide and dexamethasone (IsaRd versus Isa-VRd). This Intergroupe Francophone of Myeloma phase 3 study randomized 270 patients with NDMM that were TI, aged 65-79 years, to IsaRd versus Isa-VRd arms. The primary endpoint was a minimal residual disease (MRD) negativity rate at 10
-5 by next-generation sequencing at 18 months from randomization. Key secondary endpoints included response rates, MRD assessment rates, survival and safety. The 18-month MRD negativity rates at 10-5 were reported in 35 patients (26%, 95% confidence interval (CI) 19-34) in IsaRd versus 71 (53%, 95% CI 44-61) in Isa-VRd (odds ratio for MRD negativity 3.16, 95% CI 1.89-5.28, P < 0.0001). The MRD benefit was consistent across subgroups at 10-5 and 10-6 , and was already observed at month 12. The proportion of patients with complete response or better at 18 months was higher with Isa-VRd (58% versus 33%; P < 0.0001), as was the proportion of MRD negativity and complete response or better (37% versus 17%; P = 0.0003). At a median follow-up of 23.5 months, no difference was observed for survival times (immature data). The addition of weekly bortezomib did not significantly affect the relative dose intensity of IsaRd. Isa-VRd significantly increased MRD endpoints, including the 18-month negativity rate at 10-5 , the primary endpoint, compared with IsaRd. This study proposes Isa-VRd as a new SOC for patients with NDMM that are TI. ClinicalTrials.gov identifier: NCT04751877 ., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
24. Genomic profiling of mycosis fungoides identifies patients at high risk of disease progression.
- Author
-
Fléchon L, Arib I, Dutta AK, Hasan Bou Issa L, Sklavenitis-Pistofidis R, Tilmont R, Stewart C, Dubois R, Poulain S, Copin MC, Javed S, Nudel M, Cavalieri D, Escure G, Gower N, Chauvet P, Gazeau N, Saade C, Thiam MB, Ouelkite-Oumouchal A, Gaggero S, Cailliau É, Faiz S, Carpentier O, Duployez N, Idziorek T, Mortier L, Figeac M, Preudhomme C, Quesnel B, Mitra S, Morschhauser F, Getz G, Ghobrial IM, and Manier S
- Subjects
- Humans, Male, Female, Genomics methods, Middle Aged, Skin Neoplasms genetics, Skin Neoplasms mortality, Skin Neoplasms pathology, Mutation, Prognosis, Adult, Exome Sequencing, Aged, Risk Factors, Mycosis Fungoides genetics, Mycosis Fungoides mortality, Mycosis Fungoides diagnosis, Mycosis Fungoides pathology, Disease Progression
- Abstract
Abstract: Mycosis fungoides (MF) is the most prevalent primary cutaneous T-cell lymphoma, with an indolent or aggressive course and poor survival. The pathogenesis of MF remains unclear, and prognostic factors in the early stages are not well established. Here, we characterized the most recurrent genomic alterations using whole-exome sequencing of 67 samples from 48 patients from Lille University Hospital (France), including 18 sequential samples drawn across stages of the malignancy. Genomic data were analyzed on the Broad Institute's Terra bioinformatics platform. We found that gain7q, gain10p15.1 (IL2RA and IL15RA), del10p11.22 (ZEB1), or mutations in JUNB and TET2 are associated with high-risk disease stages. Furthermore, gain7q, gain10p15.1 (IL2RA and IL15RA), del10p11.22 (ZEB1), and del6q16.3 (TNFAIP3) are coupled with shorter survival. Del6q16.3 (TNFAIP3) was a risk factor for progression in patients at low risk. By analyzing the clonal heterogeneity and the clonal evolution of the cohort, we defined different phylogenetic pathways of the disease with acquisition of JUNB, gain10p15.1 (IL2RA and IL15RA), or del12p13.1 (CDKN1B) at progression. These results establish the genomics and clonality of MF and identify potential patients at risk of progression, independent of their clinical stage., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
25. Round Table Discussion on Optimal Clinical Trial Design in Precursor Multiple Myeloma.
- Author
-
Ghobrial IM, Gormley N, Kumar SK, Mateos MV, Bergsagel PL, Chesi M, Dhodapkar MV, Dispenzieri A, Fonseca R, Getz G, Kastritis E, Kristinsson SY, Martinez-Climent JA, Manier S, Marinac CR, Maura F, Morgan GJ, Davies FE, Nadeem O, Nuvolone M, Paiva B, O'Donnell E, Prosper F, Shah UA, Sklavenitis-Pistofidis R, Sperling AS, Vassiliou GS, Munshi NC, Castle PE, Anderson KC, and San Miguel JF
- Subjects
- Humans, Research Design, Quality of Life, Multiple Myeloma therapy, Multiple Myeloma drug therapy, Clinical Trials as Topic methods
- Abstract
Summary: While the current approach to precursor hematologic conditions is to "watch and wait," this may change with the development of therapies that are safe and extend survival or delay the onset of symptomatic disease. The goal of future therapies in precursor hematologic conditions is to improve survival and prevent or delay the development of symptomatic disease while maximizing safety. Clinical trial considerations in this field include identifying an appropriate at-risk population, safety assessments, dose selection, primary and secondary trial endpoints including surrogate endpoints, control arms, and quality-of-life metrics, all of which may enable more precise benefit-risk assessment., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
26. Progression-free survival as a surrogate endpoint for overall survival in patients with relapsed or refractory multiple myeloma.
- Author
-
Dimopoulos M, Sonneveld P, Manier S, Lam A, Roccia T, Schecter JM, Cost P, Pacaud L, Poirier A, Tremblay G, Lan T, Valluri S, and Kumar S
- Subjects
- Humans, Neoplasm Recurrence, Local mortality, Female, Male, Multiple Myeloma mortality, Multiple Myeloma therapy, Multiple Myeloma pathology, Progression-Free Survival, Randomized Controlled Trials as Topic
- Abstract
Objectives: The goal of the research was to assess the quantitative relationship between median progression-free survival (PFS) and median overall survival (OS) specifically among patients with relapsed/refractory multiple myeloma (RRMM) based on published randomized controlled trials (RCTs)., Methods: Two bibliographic databases (PubMed and Embase, 1970-2017) were systematically searched for RCTs in RRMM that reported OS and PFS, followed by an updated search of studies published between 2010 and 2022 in 3 databases (Embase, MEDLINE, and EBM Reviews, 2010-2022). The association between median PFS and median OS was assessed using the nonparametric Spearman rank and parametric Pearson correlation coefficients. Subsequently, the quantitative relationship between PFS and OS was assessed using weighted least-squares regression adjusted for covariates including age, sex, and publication year. Study arms were weighted by the number of patients in each arm., Results: A total of 31 RCTs (56 treatment arms, 10,450 patients with RRMM) were included in the analysis. The average median PFS and median OS were 7.1 months (SD 5.5) and 28.1 months (SD 11.8), respectively. The Spearman and Pearson correlation coefficients between median PFS and median OS were 0.80 (P < 0.0001) and 0.79 (P < 0.0001), respectively. In individual treatment arms of RRMM trials, each 1-month increase in median PFS was associated with a 1.72-month (95% CI 1.26-2.17) increase in median OS., Conclusion: Analysis of the relationship between PFS and OS incorporating more recent studies in RRMM further substantiates the use of PFS to predict OS in RRMM., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
27. Small myeloid subclones are present at diagnosis of multiple myeloma in patients who develop secondary myelodysplastic syndromes.
- Author
-
Escure G, Fournier E, Saade C, Issa LHB, Arib I, Tilmont R, Gazeau N, Thiam BM, Chovet M, Delforge M, Gower N, Fléchon L, Cavalieri D, Chauvet P, Nudel M, Goursaud L, Berthon C, Quesnel B, Facon T, Preudhomme C, Duployez N, and Manier S
- Subjects
- Humans, Multiple Myeloma complications, Multiple Myeloma diagnosis, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes etiology, Leukemia, Myeloid, Acute diagnosis, Neoplasms, Second Primary
- Published
- 2024
- Full Text
- View/download PDF
28. MYC dependency in GLS1 and NAMPT is a therapeutic vulnerability in multiple myeloma.
- Author
-
Hasan Bou Issa L, Fléchon L, Laine W, Ouelkdite A, Gaggero S, Cozzani A, Tilmont R, Chauvet P, Gower N, Sklavenitis-Pistofidis R, Brinster C, Thuru X, Touil Y, Quesnel B, Mitra S, Ghobrial IM, Kluza J, and Manier S
- Abstract
Multiple myeloma (MM) is an incurable hematological malignancy in which MYC alterations contribute to the malignant phenotype. Nevertheless, MYC lacks therapeutic druggability. Here, we leveraged large-scale loss-of-function screens and conducted a small molecule screen to identify genes and pathways with enhanced essentiality correlated with MYC expression. We reported a specific gene dependency in glutaminase (GLS1), essential for the viability and proliferation of MYC overexpressing cells. Conversely, the analysis of isogenic models, as well as cell lines dataset (CCLE) and patient datasets, revealed GLS1 as a non-oncogenic dependency in MYC-driven cells. We functionally delineated the differential modulation of glutamine to maintain mitochondrial function and cellular biosynthesis in MYC overexpressing cells. Furthermore, we observed that pharmaceutical inhibition of NAMPT selectively affects MYC upregulated cells. We demonstrate the effectiveness of combining GLS1 and NAMPT inhibitors, suggesting that targeting glutaminolysis and NAD synthesis may be a promising strategy to target MYC-driven MM., Competing Interests: The authors declare no conflict of interest., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
29. Prognostic impact of translocation t(14;16) in multiple myeloma according to the presence of additional genetic lesions.
- Author
-
Schavgoulidze A, Perrot A, Cazaubiel T, Leleu X, Montes L, Jacquet C, Belhadj K, Brechignac S, Frenzel L, Chalopin T, Rey P, Schiano de Collela JM, Dib M, Caillot D, Macro M, Fontan J, Buisson L, Pavageau L, Roussel M, Manier S, Mohty M, Martinet L, Avet-Loiseau H, and Corre J
- Subjects
- Humans, Prognosis, Translocation, Genetic, In Situ Hybridization, Fluorescence, Chromosomes, Human, Pair 14 genetics, Multiple Myeloma genetics, Multiple Myeloma pathology
- Published
- 2023
- Full Text
- View/download PDF
30. Real-world study of the efficacy and safety of belantamab mafodotin (GSK2857916) in relapsed or refractory multiple myeloma based on data from the nominative ATU in France: the IFM 2020-04 study.
- Author
-
Talbot A, Bobin A, Tabone L, Lambert J, Boccaccio C, Deal C, Petillon MO, Allangba O, Agape P, Arnautou P, Belkhir R, Cailleres S, Chaoui D, Chrétien ML, Decaux O, Schulmann S, Frenzel L, Gastaud L, Huart A, Hulin C, Karlin L, Laribi K, Le Calloch R, Lenain P, Macro M, Manier S, Montes L, Moreau S, Moreau P, Morel V, Norwood J, Piocelle FO, Perrot A, Pica GM, Rey P, Schmitt A, Stoppa AM, Tiab M, Touzeau C, Vidal V, Vignon M, Vincent L, Van De Wyngaert Z, Zarnitsky C, Kerbouche N, Paka P, Leleu X, Arnulf B, Avet-Loiseau H, and Du Myélome IIF
- Subjects
- Adult, Humans, Aged, Treatment Outcome, Retrospective Studies, France, Multiple Myeloma drug therapy
- Abstract
Belantamab mafodotin (BM) is an anti-BCMA antibody-drug conjugate (GSK2857916) that represents an alternative option in multiple myeloma. We sought to assess the efficacy and safety of BM in a real-world setting in patients who benefited from an early access program. We conducted an observational, retrospective, multicenter study. Eligibility criteria were treatment of relapsed or refractory multiple myeloma (RRMM) in monotherapy in adult patients who have received at least three lines of therapy previously, including at least one immunomodulatory agent (IMiD), a proteasome inhibitor (PI) and an anti-CD38 monoclonal antibody, and whose disease progressed during the last treatment period. The primary endpoint of the study is to assess the overall survival (OS). Between November 2019 and December 2020, 106 patients were treated with BM; 97 were eligible for the efficacy evaluation and 104 for safety. The median age was 66 (range, 37-82) years. High-risk cytogenetics were identified in 40.9% of patients. Fifty-five (56.7%) patients were triple-class refractory and 11 (11.3%) were penta-class refractory. The median number of prior lines of treatment was five (range, 3-12). The median number of BM cycles administered was three (range, 1-22). The overall response rate at best response was 38.1% (37/97). The median OS was 9.3 months (95% confidence interval [CI]: 5.9-15.3), and median progression-free survival was 3.5 months (95% CI: 1.9-4.7). The median duration of response was 9 months (range, 4.65-10.4). Treatment was delayed for 55 (52.9%) patients including 36.5% for treatment-related toxicity. Ophthalmic adverse events, mainly grade ≤2, were the most common toxicity (48%). The occurrence of keratopathy was 37.5%. Overall, our data are concordant with the results from DREAMM-2 in terms of efficacy and safety on a non-biased population.
- Published
- 2023
- Full Text
- View/download PDF
31. Elranatamab in relapsed or refractory multiple myeloma: phase 2 MagnetisMM-3 trial results.
- Author
-
Lesokhin AM, Tomasson MH, Arnulf B, Bahlis NJ, Miles Prince H, Niesvizky R, Rodrίguez-Otero P, Martinez-Lopez J, Koehne G, Touzeau C, Jethava Y, Quach H, Depaus J, Yokoyama H, Gabayan AE, Stevens DA, Nooka AK, Manier S, Raje N, Iida S, Raab MS, Searle E, Leip E, Sullivan ST, Conte U, Elmeliegy M, Czibere A, Viqueira A, and Mohty M
- Subjects
- Humans, B-Cell Maturation Antigen, Progression-Free Survival, Remission Induction, Multiple Myeloma drug therapy
- Abstract
Elranatamab is a humanized B-cell maturation antigen (BCMA)-CD3 bispecific antibody. In the ongoing phase 2 MagnetisMM-3 trial, patients with relapsed or refractory multiple myeloma received subcutaneous elranatamab once weekly after two step-up priming doses. After six cycles, persistent responders switched to biweekly dosing. Results from cohort A, which enrolled patients without prior BCMA-directed therapy (n = 123) are reported. The primary endpoint of confirmed objective response rate (ORR) by blinded independent central review was met with an ORR of 61.0% (75/123); 35.0% ≥complete response. Fifty responders switched to biweekly dosing, and 40 (80.0%) improved or maintained their response for ≥6 months. With a median follow-up of 14.7 months, median duration of response, progression-free survival and overall survival (secondary endpoints) have not been reached. Fifteen-month rates were 71.5%, 50.9% and 56.7%, respectively. Common adverse events (any grade; grade 3-4) included infections (69.9%, 39.8%), cytokine release syndrome (57.7%, 0%), anemia (48.8%, 37.4%), and neutropenia (48.8%, 48.8%). With biweekly dosing, grade 3-4 adverse events decreased from 58.6% to 46.6%. Elranatamab induced deep and durable responses with a manageable safety profile. Switching to biweekly dosing may improve long-term safety without compromising efficacy. ClinicalTrials.gov identifier: NCT04649359 ., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
32. Measurable Residual Disease Testing in Multiple Myeloma Routine Clinical Practice: A Modified Delphi Study.
- Author
-
Ramasamy K, Avet-Loiseau H, Hveding Blimark C, Delforge M, Gay F, Manier S, Martinez-Lopez J, Mateos MV, Mohty M, van de Donk NWCJ, and Weisel K
- Abstract
We used a modified Delphi approach to establish areas of consensus and nonconsensus regarding the utility of determining measurable residual disease (MRD) to assess multiple myeloma (MM) treatment response, which may inform disease management and design of future clinical trials. This modified Delphi study incorporated 2 iterative rounds of surveys to evaluate the opinions of an expert panel of 61 practicing hematological oncologists from across 14 countries in Europe concerning the use of MRD testing in MM management. Survey 1 assessed experts' opinions on MRD testing in different clinical situations and associated challenges. Survey 2 focused on the lack of consensus areas identified in survey 1. Consensus to an individual question was defined a priori as 75% agreement or disagreement by the panel. From the 2 rounds of surveys, the experts reached consensus agreement that MRD testing should be performed in newly diagnosed or relapsed patients who achieved complete response (CR) or better after transplantation. In transplant-ineligible patients, experts recommended MRD testing in those who are ≤70 years old and in CR. If a patient was previously positive on positron-emission tomography and computed tomography (PET/CT), both MRD and PET/CT should be assessed at CR. MRD testing should be performed ≤6 months after transplantation and every 6-12 months in continuously treated patients in CR. There was no consensus on making treatment decisions based on MRD status. MRD testing is an important component of clinical management in MM. Additional data will further clarify the role of MRD in guiding treatment decisions., Competing Interests: KR: Has served as an advisor for and received honoraria from Janssen, Adaptive Biotechnologies, Amgen, Takeda, Abbvie, Oncopeptides, Celgene – Bristol Myers Squibb, Pfizer, and GSK. Has received grant funding from Janssen, Amgen, Takeda, GSK and Celgene – Bristol Myers Squibb. HA-L: Has served as an advisor for and received honoraria from Amgen, Bristol Myers Squibb, Celgene, Janssen, Sanofi, GSK, Takeda, and Adaptive Biotechnologies. CHB: Has served as an advisor for and received honoraria from Amgen, Bristol Myers Squibb, Takeda, Sanofi, GSK, and Adaptive Biotechnologies. MD: Has served as an advisor for and received honoraria from Amgen, Celgene – Bristol Myers Squibb, GSK, Janssen, Sanofi, and Takeda. FG: Has served as an advisor for Janssen, Amgen, Celgene – Bristol Myers Squibb, Adaptive Biotechnologies, Roche, Abbvie, GSK, Takeda, Bluebird Bio, Oncopeptides, Pfizer, and Sanofi. Has received honoraria from Janssen, Amgen, Celgene – Bristol Myers Squibb, GSK, Takeda, and Sanofi. SM: Has served as an advisor for Abbvie, Adaptive Biotechnologies, Amgen, Celgene – Bristol Myers Squibb, GSK, Janssen, Oncopeptides, Pfizer, Regeneron, Roche, Sanofi, and Takeda. JM-L: Has served as an advisor for and received honoraria from Amgen, Bristol Myers Squibb, Celgene, Janssen, Sanofi, GSK, Incyte, Roche, Pfizer, Novartis and Adaptive Biotechnologies. MVM: Has served as an advisor for and received honoraria from Janssen, Celgene – Bristol Myers Squibb, Takeda, Amgen, Adaptive Biotechnologies, Oncopeptides, Sanofi, Roche, Regeneron, and Pfizer. MM: Has received honoraria from Amgen, Astellas, Bristol Myers Squibb, Celgene, Gilead, Janssen, Jazz, Takeda, Novartis, Pfizer, Sanofi, and Adaptive Biotechnologies. Has received research funding from Celgene, Janssen, Jazz, and Sanofi. NWCJvdD: Has served as an advisor for Janssen Pharmaceuticals, Amgen, Adaptive Biotechnologies, Celgene, Bristol Myers Squibb, Novartis, Roche, Takeda, Bayer, and Servier. Has received grant funding from Janssen Pharmaceuticals, Amgen, Celgene, Cellectis, and Bristol Myers Squibb. KW: Has served as an advisor for Amgen, Adaptive Biotechnologies, Bristol Myers Squibb, Celgene, GSK, Janssen, Karyopharm, Oncopeptides, Sanofi, and Takeda. Has received honoraria from Amgen, Adaptive Biotechnologies, Bristol Myers Squibb, Celgene, GSK, Janssen, Karyopharm, Oncopeptides, Sanofi, Takeda, and Roche. Has received grant funding from Amgen, Janssen, Celgene, and Sanofi (to the institution)., (Copyright © 2023 the Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the European Hematology Association.)
- Published
- 2023
- Full Text
- View/download PDF
33. Difficult-to-treat patients with relapsed/refractory multiple myeloma: A review of clinical trial results.
- Author
-
Raab MS, Zamagni E, Manier S, Rodriguez-Otero P, Schjesvold F, and Broijl A
- Abstract
Overall outcomes for multiple myeloma have improved due to the availability of new therapies, but patients with relapsed/refractory multiple myeloma harbouring certain factors continue to pose a therapeutic challenge. These challenging features include high-risk cytogenetics, renal impairment, patient characteristics such as age and frailty, and extramedullary disease. Prior refractory status and number of prior lines add further complexity to the treatment of these patients. While newer regimens are available and have suggested efficacy in these patient populations through subgroup analyses, differences in trial definitions and cut-offs make meaningful comparisons difficult. This review aims to examine the available clinical trial data for patients with high-risk cytogenetics, renal impairment, age and frailty and extramedullary disease., Competing Interests: M‐SR: honoraria – BMS, Janssen; advisory role – Amgen, BMS, Janssen, Novartis, Sanofi, Takeda; consultant – Amgen, BMS, Janssen, Novartis, Sanofi, Takeda; research support – BMS, Janssen, Novartis, Sanofi. EZ: honoraria – Amgen, BMS, GSK, Janssen, Pfizer, Roche, Sanofi, Takeda; advisory role – Amgen, BMS, GSK, Janssen, Pfizer, Roche, Sanofi, Takeda. SM: advisory role – AbbVie, Adaptive Biotechnology, Amgen, Celgene/BMS, GlaxoSmithKline, Janssen, Novartis, Oncopeptide, Regeneron, Roche, Sanofi, Takeda; research support – AbbVie, Adaptive Biotechnology, Amgen, Celgene/BMS, GlaxoSmithKline, Janssen, Novartis, Oncopeptide, Regeneron, Roche, Sanofi, Takeda. PR‐O: honoraria – Amgen, BMS, GSK, Janssen, Regeneron, Sanofi, Takeda; advisory role – Abbvie, BMS, GSK, Janssen, Kite Pharma, Oncopeptides, Pfizer, Sanofi, Takeda; consultant – Celgene‐BMS, GSK, Pfizer. FS: honoraria – Abbvie, Amgen, BMS, Daiichi‐Sankyo, GSK, Janssen, Novartis, Oncopeptides, Pfizer, Sanofi, SkyliteDX, Takeda; advisory role – Abbvie, Celgene, GSK, Janssen, Oncopeptides, Sanofi, Takeda; research support – Celgene, GSK, Janssen, Oncopeptides, Sanofi, Targovax. AB: honoraria – Amgen, BMS, Janssen, Sanofi, Takeda; advisory role – Amgen, BMS, Janssen, Sanofi, Takeda., (© 2023 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
34. Current Advances in Multiple Myeloma: A Post International Myeloma Society (IMS 2022) Round Table Debate by the International Academy for Clinical Hematology (IACH).
- Author
-
Bahlis NJ, Costa LJ, Facon T, Harousseau JL, Manier S, Perrot A, Touzeau C, and Mohty M
- Abstract
This round table discussion organized by the International Academy for Clinical Hematology (IACH) was dedicated to the 19th annual meeting of the International Myeloma Society (IMS), which was held in Los Angeles between the 25th and 27th August 2022. After some key meetings of the discipline of the field of clinical hematology, the IACH organizes regular round table discussion in order to summarize the flow of information and get the opinion of a panel of experts and the key take-home messages. As part of this discussion, the panellists debated 6 key topics: disease monitoring, management of high-risk multiple myeloma (MM), induction for newly-diagnosed MM, management of relapsed MM, immune reconstitution, and vaccination and cellular therapy in MM., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
35. Biallelic deletion of 1p32 defines ultra-high-risk myeloma, but monoallelic del(1p32) remains a strong prognostic factor.
- Author
-
Schavgoulidze A, Talbot A, Perrot A, Cazaubiel T, Leleu X, Manier S, Buisson L, Mahéo S, Do Souto Ferreira L, Pavageau L, Hulin C, Marolleau JP, Voillat L, Belhadj K, Divoux M, Slama B, Brechignac S, Macro M, Stoppa AM, Sanhes L, Orsini-Piocelle F, Fontan J, Chretien ML, Demarquette H, Mohty M, Avet-Loiseau H, and Corre J
- Subjects
- Humans, Prognosis, Chromosome Aberrations, In Situ Hybridization, Fluorescence, Progression-Free Survival, Multiple Myeloma diagnosis, Multiple Myeloma genetics
- Abstract
Cytogenetic abnormalities (CAs) are known to be the preponderant prognostic factor in multiple myeloma. Our team has recently developed a prognostic score based on 6 CAs, with which del(1p32) appears to be the second worst abnormality after del(17p). This study aimed to confirm the adverse effect of 1p32 deletion in patients with newly diagnosed multiple myeloma (NDMM). Among 2551 patients with newly diagnosed multiple myeloma, 11% were harboring del(1p32). Their overall survival (OS) was significantly inferior compared with patients without del(1p32) (median OS: 49 months vs 124 months). Likewise, progression-free survival was significantly shorter. More importantly, biallelic del(1p32) conferred a dramatically poorer prognosis than a monoallelic del(1p32) (median OS: 25 months vs 60 months). As expected, the OS of patients with del(1p32) significantly decreased when this abnormality was associated with other high-risk CAs [del(17p), t(4;14), or gain(1q)]. In the multivariate analysis, del(1p32) appeared as a negative prognostic factor; after adjustment for age and treatment, the risk of progression was 1.3 times higher among patients harboring del(1p32), and the risk of death was 1.9 times higher. At the dawn of risk-adapted treatment strategies, we have confirmed the adverse effect of del(1p32) in multiple myeloma and the relevance of its assessment at diagnosis., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
36. Clinical characteristics and outcome of 318 families with familial monoclonal gammopathy: A multicenter Intergroupe Francophone du Myélome study.
- Author
-
Dumontet C, Demangel D, Galia P, Karlin L, Roche L, Fauvernier M, Golfier C, Laude MC, Leleu X, Rodon P, Roussel M, Azaïs I, Doyen C, Slama B, Manier S, Decaux O, Pertesi M, Beaumont M, Caillot D, Boyle EM, Cliquennois M, Cony-Makhoul P, Doncker AV, Dorvaux V, Petillon MO, Fontan J, Hivert B, Leduc I, Leyronnas C, Macro M, Maigre M, Mariette C, Mineur P, Rigaudeau S, Royer B, Vincent L, Mckay J, Perrial E, and Garderet L
- Subjects
- Child, Humans, Prognosis, Chromosome Aberrations, Monoclonal Gammopathy of Undetermined Significance diagnosis, Paraproteinemias genetics, Paraproteinemias complications, Multiple Myeloma pathology
- Abstract
Familial forms of monoclonal gammopathy, defined as multiple myeloma (MM) or Monoclonal Gammopathy of Undetermined Significance (MGUS), are relatively infrequent and most series reported in the literature describe a limited number of families. MM rarely occurs in a familial context. MGUS is observed much more commonly, which can in some cases evolve toward full-blown MM. Although recurrent cytogenetic abnormalities have been described in tumor cells of sporadic cases of MM, the pathogenesis of familial MM remains largely unexplained. In order to identify genetic factors predisposing to familial monoclonal gammopathy, the Intergroupe Francophone du Myélome identified 318 families with at least two confirmed cases of monoclonal gammopathy. There were 169 families with parent/child pairs and 164 families with cases in at least two siblings, compatible with an autosomal transmission. These familial cases were compared with sporadic cases who were matched for age at diagnosis, sex and immunoglobulin isotype, with 10 sporadic cases for each familial case. The gender distribution, age and immunoglobulin subtypes of familial cases were unremarkable in comparison to sporadic cases. With a median follow-up of 7.4 years after diagnosis, the percentage of MGUS cases having evolved to MM was 3%. The median overall survival of the 148 familial MM cases was longer than that of matched sporadic cases, with projected values of 7.6 and 16.1 years in patients older and younger than 65 years, respectively. These data suggest that familial cases of monoclonal gammopathy are similar to sporadic cases in terms of clinical presentation and carry a better prognosis., (© 2023 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
37. Immune biomarkers of response to immunotherapy in patients with high-risk smoldering myeloma.
- Author
-
Sklavenitis-Pistofidis R, Aranha MP, Redd RA, Baginska J, Haradhvala NJ, Hallisey M, Dutta AK, Savell A, Varmeh S, Heilpern-Mallory D, Ujwary S, Zavidij O, Aguet F, Su NK, Lightbody ED, Bustoros M, Tahri S, Mouhieddine TH, Wu T, Flechon L, Anand S, Rosenblatt JM, Zonder J, Vredenburgh JJ, Boruchov A, Bhutani M, Usmani SZ, Matous J, Yee AJ, Jakubowiak A, Laubach J, Manier S, Nadeem O, Richardson P, Badros AZ, Mateos MV, Trippa L, Getz G, and Ghobrial IM
- Subjects
- Humans, Biomarkers, Disease Progression, Immunologic Factors, Immunotherapy, Lenalidomide adverse effects, Clinical Trials, Phase II as Topic, Multiple Myeloma drug therapy, Smoldering Multiple Myeloma therapy
- Abstract
Patients with smoldering multiple myeloma (SMM) are observed until progression, but early treatment may improve outcomes. We conducted a phase II trial of elotuzumab, lenalidomide, and dexamethasone (EloLenDex) in patients with high-risk SMM and performed single-cell RNA and T cell receptor (TCR) sequencing on 149 bone marrow (BM) and peripheral blood (PB) samples from patients and healthy donors (HDs). We find that early treatment with EloLenDex is safe and effective and provide a comprehensive characterization of alterations in immune cell composition and TCR repertoire diversity in patients. We show that the similarity of a patient's immune cell composition to that of HDs may have prognostic relevance at diagnosis and after treatment and that the abundance of granzyme K (GZMK)
+ CD8+ effector memory T (TEM) cells may be associated with treatment response. Last, we uncover similarities between immune alterations observed in the BM and PB, suggesting that PB-based immune profiling may have diagnostic and prognostic utility., Competing Interests: Declaration of interests N.J.H. is a consultant for Constellation Pharmaceuticals. F.A. is an employee of Illumina Inc. O.Z. is an employee of Ikena Oncology and a stockholder in Ikena Oncology and Morphosys AG. G.G. receives research funds from IBM and Pharmacyclics and is an inventor on patent applications filed by the Broad Institute related to MSMuTect, MSMutSig, POLYSOLVER, SignatureAnalyzer-GPU, and MSIDetect. He is also a founder and consultant of and holds privately held equity in Scorpion Therapeutics. I.M.G. has a consulting or advisory role with AbbVie, Adaptive, Amgen, Aptitude Health, Bristol Myers Squibb, GlaxoSmithKline, Huron Consulting, Janssen, Menarini Silicon Biosystems, Oncopeptides, Pfizer, Sanofi, Sognef, Takeda, The Binding Site, and Window Therapeutics and has received speaker fees from Vor Biopharma and Veeva Systems, Inc., and her spouse is the CMO and equity holder of Disc Medicine. S.M. has a consulting role with Abbvie, Adaptive Biotechnology, Amgen, Celgene/BMS, GlaxoSmithKline, Janssen, Novartis, Oncopeptides, Regeneron, Roche, and Takeda and has received research funding from Abbvie, Adaptive Biotechnology, Amgen, Celgene/BMS, GlaxoSmithKline, Janssen, Novartis, Oncopeptides, Regeneron, Roche, and Takeda. A.J.Y. has a consulting role with Adaptive Biotechnologies, Amgen, BMS, Celgene, GSK, Janssen, Karyopharm, Oncopeptides, Sanofi, and Takeda and has received research funding from Amgen, Janssen, and Takeda. M.B. is a consultant for Sanofi, Genzyme, and Janssen and has received research funding from MedImmune, Janssen, Legend Biotech, Amgen, Celularity, Bristol Myers Squibb, Celgene, Bluebird bio, Millennium, Takeda, Cerecor (currently Avalo Therapeutics), and C4 Therapeutics. M.B has an advisory role and received honoraria from Bristol Myers Squibb, Takeda, Janssen, and Menarini. T.H.M. received advisory board fees from Legend Biotech. R.S.-P., G.G., and I.M.G. are co-inventors on a patent application related to this work (PCT/US22/74839)., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
38. All-oral triplet combination of ixazomib, lenalidomide, and dexamethasone in newly diagnosed transplant-eligible multiple myeloma patients: final results of the phase II IFM 2013-06 study.
- Author
-
Touzeau C, Perrot A, Roussel M, Karlin L, Benboubker L, Jacquet C, Mohty M, Facon T, Manier S, Chretien ML, Tiab M, Hulin C, Leleu X, Loiseau HA, Dejoie T, Planche L, Attal M, and Moreau P
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Boron Compounds therapeutic use, Dexamethasone adverse effects, Glycine analogs & derivatives, Humans, Lenalidomide therapeutic use, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy
- Published
- 2022
- Full Text
- View/download PDF
39. Bortezomib and high-dose melphalan conditioning regimen in frontline multiple myeloma: an IFM randomized phase 3 study.
- Author
-
Roussel M, Lauwers-Cances V, Macro M, Leleu X, Royer B, Hulin C, Karlin L, Perrot A, Touzeau C, Chrétien ML, Rigaudeau S, Dib M, Nicolas-Virelizier E, Escoffre-Barbe M, Belhadj K, Mariette C, Stoppa AM, Araujo C, Doyen C, Fontan J, Kolb B, Garderet L, Brechignac S, Malfuson JV, Jaccard A, Lenain P, Borel C, Hebraud B, Benbrahim O, Dorvaux V, Manier S, Augeul-Meunier K, Vekemans MC, Randriamalala E, Chaoui D, Caers J, Chaleteix C, Benboubker L, Vincent L, Glaisner S, Zunic P, Slama B, Eveillard JR, Humbrecht-Kraut C, Morel V, Mineur P, Eisenmann JC, Demarquette H, Richez V, Vignon M, Caillot D, Facon T, Moreau P, Colin AL, Olivier P, Wuilleme S, Avet-Loiseau H, Corre J, and Attal M
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib adverse effects, Humans, Transplantation, Autologous, Melphalan adverse effects, Multiple Myeloma drug therapy, Multiple Myeloma etiology
- Abstract
High-dose melphalan (HDM) and transplantation are recommended for eligible patients with multiple myeloma. No other conditioning regimen has proven to be more effective and/or safer. We previously reported in a phase 2 study that bortezomib can safely and effectively be combined with HDM (Bor-HDM), with a 32% complete response (CR) rate after transplantation. These data supported a randomized phase 3 trial. Randomization was stratified according to risk and response to induction: 300 patients were enrolled, and 154 were allocated to the experimental arm (ie, arm A) with bortezomib (1 mg/m2 intravenously [IV]) on days -6, -3, +1, and +4 and melphalan (200 mg/m2 IV) on day -2. The control arm (ie, arm B) consisted of HDM alone (200 mg/m2 IV). There were no differences in stringent CR + CR rates at day 60 posttransplant (primary end point): 22.1% in arm A vs 20.5% in arm B (P = .844). There were also no differences in undetectable minimum residual disease rates: 41.3% vs 39.4% (P = .864). Median progression-free survival was 34.0 months for arm A vs 29.6 months for arm B (adjusted HR, 0.82; 95% CI, 0.61-1.13; P = .244). The estimated 3-year overall survival was 89.5% in both arms (hazard ratio, 1.28; 95% CI, 0.62-2.64; P = .374). Sixty-nine serious adverse events occurred in 18.7% of Bor-HDM-treated patients (vs 13.1% in HDM-treated patients). The proportion of grade 3/4 AEs was similar within the 2 groups (72.0% vs 73.1%), mainly (as expected) blood and gastrointestinal disorders; 4% of patients reported grade 3/4 or painful peripheral neuropathy in arm A (vs 1.5% in arm B). In this randomized phase 3 study, a conditioning regimen with Bor-HDM did not improve efficacy end points or outcomes compared with HDM alone. The original trial was registered at www.clinicaltrials.gov as #NCT02197221., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
40. Primary plasma cell leukemias displaying t(11;14) have specific genomic, transcriptional, and clinical features.
- Author
-
Cazaubiel T, Leleu X, Perrot A, Manier S, Buisson L, Maheo S, Do Souto Ferreira L, Lannes R, Pavageau L, Hulin C, Marolleau JP, Voillat L, Belhadj K, Divoux M, Slama B, Brechignac S, Macro M, Stoppa AM, Sanhes L, Orsini-Piocelle F, Fontan J, Chretien ML, Demarquette H, Mohty M, Schavgoulidze A, Avet-Loiseau H, and Corre J
- Subjects
- Chromosome Aberrations, Genomics, Humans, Prognosis, Transcriptome, Leukemia, Plasma Cell diagnosis, Multiple Myeloma genetics
- Abstract
Primary plasma cell leukemia (pPCL) is an aggressive form of multiple myeloma (MM) that has not benefited from recent therapeutic advances in the field. Because it is very rare and heterogeneous, it remains poorly understood at the molecular level. To address this issue, we performed DNA and RNA sequencing of sorted plasma cells from a large cohort of 90 newly diagnosed pPCL and compared with MM. We observed that pPCL presents a specific genomic landscape with a high prevalence of t(11;14) (about half) and high-risk genomic features such as del(17p), gain 1q, and del(1p32). In addition, pPCL displays a specific transcriptome when compared with MM. We then wanted to characterize specifically pPCL with t(11;14). We observed that this subentity displayed significantly fewer adverse cytogenetic abnormalities. This translated into better overall survival when compared with pPCL without t(11;14) (39.2 months vs 17.9 months, P = .002). Finally, pPCL with t(11;14) displayed a specific transcriptome, including differential expression of BCL2 family members. This study is the largest series of patients with pPCL reported so far., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
41. Epidemiological landscape of young patients with multiple myeloma diagnosed before 40 years of age: the French experience.
- Author
-
Caulier A, Roussel M, Morel P, Lombion N, Branco B, Galtier J, Hulin C, Perrot A, Richez V, Michaud AV, Touzeau C, Doyen C, Mariette C, Caillot D, Harel S, Lenain P, Ivanoff S, Fontan J, Stoppa AM, Manier S, Garderet L, Leleu X, Marolleau JP, Arnulf B, Avet-Loiseau H, and Royer B
- Subjects
- Adult, Age Factors, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Female, Follow-Up Studies, France epidemiology, Hematopoietic Stem Cell Transplantation, Humans, Male, Multiple Myeloma therapy, Progression-Free Survival, Transplantation, Autologous, Treatment Outcome, Young Adult, Multiple Myeloma epidemiology
- Abstract
Multiple myeloma (MM) is rare in young patients, especially before age 40 years at diagnosis, representing <2% of all patients with MM. Little is known about the disease characteristics and prognosis of these patients. In this study, we examined 214 patients diagnosed with MM at age ≤40 years over 15 years, in the era of modern treatments. Among them, 189 patients had symptomatic MM. Disease characteristics were similar to older patients: 35% had anemia, 17% had renal impairment, and 13% had hypercalcemia. The staging was ISS-1 in 52.4%, ISS-2 in 27.5%, and ISS-3 in 20.1%. Overall, 18% of patients had high-risk cytogenetics [del 17p and/or t(4;14)]. Ninety percent of patients received intensive chemotherapy followed by autologous stem cell transplant, and 25% of patients had allogeneic stem cell transplant predominantly at time of relapse. The median follow-up was 76 months, the estimated median overall survival was 14.5 years, and the median progression free-survival was 41 months. In multivariate analysis, bone lesions (hazard ratio [HR], 3.95; P = .01), high ISS score (HR, 2.14; P = .03), and high-risk cytogenetics (HR, 4.54; P < .0001) were significant risk factors for poor outcomes. Among predefined time-dependent covariables, onset of progression (HR, 13.2; P < .0001) significantly shortened overall survival. At 5 years, relative survival compared with same age- and sex-matched individuals was 83.5%, and estimated standardized mortality ratio was 69.9 (95% confidence interval, 52.7-91.1), confirming that MM dramatically shortens the survival of young patients despite an extended survival after diagnosis., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
42. Longitudinal study of AMH variations in 122 Adolescents and Young Adults (AYA) and non-AYA lymphoma patients to evaluate the chemo-induced ovarian toxicity to further personalise fertility preservation counselling.
- Author
-
Decanter C, Delepine J, Behal H, Manier S, Bruno B, Barbatti M, Robin C, Labreuche J, Morschhauser F, and Pigny P
- Subjects
- Adolescent, Adult, Anti-Mullerian Hormone, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bleomycin therapeutic use, Counseling, Dacarbazine therapeutic use, Doxorubicin therapeutic use, Female, Humans, Longitudinal Studies, Prospective Studies, Vinblastine therapeutic use, Young Adult, Fertility Preservation, Hodgkin Disease drug therapy
- Abstract
Study Question: What is the influence of age and chemotherapy regimen on the longitudinal blood anti-Müllerian hormone (AMH) variations in a large series of adolescents and young adult (AYA) (15-24 years old) and non-AYA (25-35 years old) lymphoma patients?, Summary Answer: In case of alkylating regimen treatment, there was a deep and sustained follicular depletion in AYA as well as non-AYA patients; however in both groups, the ovarian toxicity was extremely low in cases of non-alkylating treatments., What Is Known Already: AMH is now well-recognised to be a real-time indicator of ovarian follicular depletion and recovery in women treated by chemotherapy. Its longitudinal variations may discriminate between highly and minimally toxic protocols regarding ovarian function. It has been shown, in different cancer types, that age, type of chemotherapy regimen and pre-treatment AMH levels are the main predictors of ovarian recovery. Large studies on longitudinal AMH variations under chemotherapy in lymphoma patients are few but can provide the opportunity to assess the degree of follicle loss at a young age., Study Design, Size, Duration: This prospective cohort study was conducted in the Fertility Observatory of the Lille University Hospital. Data were collected between 2007 and 2016. Non-Hodgkin or Hodgkin lymphoma patients (n = 122) between 15 and 35 years old were prospectively recruited before commencing chemotherapy. Patients were treated either by a non-alkylating protocol (ABVD group; n = 67) or by an alkylating regimen (alkylating group; n = 55)., Participants/materials, Setting, Methods: Serial AMH measurements were performed at baseline (AMH0), 15 days after the start of chemotherapy (AMH1), 15 days before the last chemotherapy cycle (AMH2), and at time 3, 6, 9, 12, 18 and 24 months from the end of chemotherapy. The whole study population was divided into two groups according to age: AYA (15-24; n = 65) and non-AYA (25-35; n = 57). All patients received a once monthly GnRH agonist injection during the whole treatment period. A linear mixed model was used to account for the repeated measures of single patients., Main Results and the Role of Chance: At baseline, non-AYA patients had higher BMI and lower AMH levels than AYA patients. All AYA and non-AYA patients having received ABVD protocols had regular cycles at 12 months of follow-up. In case of alkylating regimens, amenorrhoea was more frequent in non-AYA patients than in AYA patients at 12 months (37% vs 4%, P = 0.011) and at 24 months (24% vs 4%, P = 0.045). We distinguished a similar depletion phase from AMH0 to AMH2 between ABVD and alkylating groups but significantly different recovery phases from AMH2 to AMH + 24 months. AMH recovery was fast and complete in case of ABVD protocols whatever the age: AMH reached pre-treatment values as soon as the 6th month of follow-up in the AYA group (mean (95% CI) in log AMH M0 vs M6: 3.07 (2.86 to 3.27) vs 3.05 (2.78 to 3.31), P = 1.00) and in the non-AYA group (mean (95% CI) in log AMH M0 vs M6: 2.73 (2.40 to 3.05) vs 2.47 (2.21 to 2.74), P = 1.00). In contrast, no patients from the alkylating group returned to pre-treatment AMH values whatever the age of patients (AYA or non-AYA). Moreover, none of the AMH values post-chemotherapy in the non-AYA group were significantly different from AMH2. Conversely in the AYA group, AMH levels from 6 months (mean (95% CI) in log AMH: 1.79 (1.47 to 2.11), P < 0.001) to 24 months (mean (95% CI) in log AMH: 2.16 (1.80 to 2.52), P ≤ 0.001) were significantly higher than AMH2 (mean (95% CI) in log AMH: 1.13 (0.89 to 1.38)). Considering the whole study population (AYA and non-AYA), pre-treatment AMH levels influenced the pattern of the AMH variation both in alkylating and ABVD protocols (interaction P-value = 0.005 and 0.043, respectively). Likewise, age was significantly associated with the pattern of the recovery phase but only in the alkylating group (interaction P-value =0.001). BMI had no influence on the AMH recovery phase whatever the protocol (interaction P-value = 0.98 in alkylating group, 0.72 in ABVD group)., Limitations, Reasons for Caution: There was a large disparity in subtypes of protocols in the alkylating group. The average duration of chemotherapy for patients treated with alkylating protocols was longer than that for patients treated with ABVD., Wider Implications of the Findings: These results make it possible to develop strategies for fertility preservation according to age and type of protocol in a large series of young lymphoma patients. In addition, it was confirmed that young age does not protect against ovarian damage caused by alkylating agents., Study Funding/competing Interest(s): This work was supported by Agence Régionale de Santé Hauts de France and Agence Onco Hauts-de-France who provided finances for AMH dosages (n° DOS/SDES/AR/FIR/2019/282). There are no competing interests., Trial Registration Number: DC-2008-642 and CNIL DEC2015-112., (© The Author(s) 2021. Published by Oxford University Press on behalf of European Society of Human Reproduction and Embryology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
43. Effective anti-BCMA retreatment in multiple myeloma.
- Author
-
Gazeau N, Beauvais D, Yakoub-Agha I, Mitra S, Campbell TB, Facon T, and Manier S
- Subjects
- B-Cell Maturation Antigen, Humans, Retreatment, T-Lymphocytes, Tumor Microenvironment, Antibodies, Bispecific therapeutic use, Multiple Myeloma drug therapy
- Abstract
The recent emergence of anti-B-cell maturation antigen (BCMA) therapies holds great promise in multiple myeloma (MM). These include chimeric antigen receptor (CAR) T cells, bispecific antibodies, and antibody-drug conjugates. Their development in clinical trials and further approval are changing the strategy for treating MM. Considering that a cure has not been reached, a central question in the coming years will be the possibility of using these therapies sequentially. Here, we report 2 cases of the serial use of anti-BCMA therapies with parallel monitoring of BCMA expression and anti-CAR antibodies. We further discuss recent data from clinical studies that have informed us about the different mechanisms of resistance to anti-BCMA therapies, including antigen escape, BCMA shedding, anti-drug antibodies, T-cell exhaustion, and the emergence of an immunosuppressive microenvironment. This knowledge will be essential to help guide the strategy of serial treatments with anti-BCMA therapies., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
44. Up-front carfilzomib, lenalidomide, and dexamethasone with transplant for patients with multiple myeloma: the IFM KRd final results.
- Author
-
Roussel M, Lauwers-Cances V, Wuilleme S, Belhadj K, Manier S, Garderet L, Escoffre-Barbe M, Mariette C, Benboubker L, Caillot D, Sonntag C, Touzeau C, Dupuis J, Moreau P, Leleu X, Facon T, Hébraud B, Corre J, and Attal M
- Subjects
- Combined Modality Therapy, Dexamethasone adverse effects, Disease-Free Survival, Female, Humans, Kaplan-Meier Estimate, Lenalidomide adverse effects, Male, Middle Aged, Neoplasm, Residual pathology, Oligopeptides adverse effects, Survival Analysis, Treatment Outcome, Dexamethasone therapeutic use, Hematopoietic Stem Cell Transplantation, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Oligopeptides therapeutic use
- Abstract
Bortezomib, lenalidomide, and dexamethasone plus transplant is a standard of care for eligible patients with multiple myeloma. Because responses can deepen with time, regimens with longer and more potent induction/consolidation phases are needed. In this phase 2 study, patients received eight 28-day cycles of carfilzomib (K) 20/36 mg/m2 (days 1-2, 8-9, 15-16), lenalidomide (R) 25 mg (days 1-21), and dexamethasone (d) 20 mg (days 1-2, 8-9, 15-16, 22-23). All patients proceeded to transplant after 4 cycles and received 1 year of lenalidomide maintenance (10 mg, days 1-21). The primary objective was stringent complete response at the completion of consolidation. Overall, 48 patients were screened and 46 enrolled; 21% had adverse cytogenetics. Among 42 evaluable patients after consolidation, 26 were in stringent complete response (CR; 61.9%), 27 were at least in CR (64.3%): 92.6% had undetectable minimal residual disease according to flow cytometry (≥2.5 × 10-5) and 63.0% according to next-generation sequencing (10-6). Median time to CR was 10.6 months. According to multiparametric flow cytometry and next-generation sequencing, 69.0% and 66.7% of patients, respectively, had undetectable minimal residual disease at some point. With a median follow-up of 60.5 months, 21 patients progressed, and 10 died (7 of multiple myeloma). Median progression-free survival was 56.4 months. There were no KRd-related deaths. Four patients discontinued the program due to toxicities; 56 serious adverse events were reported in 31 patients, including 8 cardiovascular events (2 heart failures, 5 pulmonary embolisms or deep vein thrombosis). Common grade 3/4 adverse events were hematologic (74%) and infectious (22%). In summary, 8 cycles of KRd produce fast and deep responses in transplant-eligible patients with newly diagnosed multiple myeloma. The safety profile is acceptable, but cardiovascular adverse events should be closely monitored. This clinical trial is registered at www.clinicaltrials.gov as #NCT02405364., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
45. Progression signature underlies clonal evolution and dissemination of multiple myeloma.
- Author
-
Shen YJ, Mishima Y, Shi J, Sklavenitis-Pistofidis R, Redd RA, Moschetta M, Manier S, Roccaro AM, Sacco A, Tai YT, Mercier F, Kawano Y, Su NK, Berrios B, Doench JG, Root DE, Michor F, Scadden DT, and Ghobrial IM
- Subjects
- Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adaptor Proteins, Signal Transducing genetics, Animals, Apoptosis, Biomarkers, Tumor genetics, Bone Marrow metabolism, Bone Marrow pathology, CRISPR-Cas Systems, Cell Adhesion, Cell Movement, Cell Proliferation, Clonal Evolution, Disease Progression, Female, HMGA1a Protein antagonists & inhibitors, HMGA1a Protein genetics, Humans, Mice, Mice, SCID, Multiple Myeloma genetics, Multiple Myeloma metabolism, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local metabolism, Prognosis, RNA-Binding Proteins antagonists & inhibitors, RNA-Binding Proteins genetics, Survival Rate, Tumor Cells, Cultured, Adaptor Proteins, Signal Transducing metabolism, Biomarkers, Tumor metabolism, Disease Models, Animal, Gene Expression Regulation, Neoplastic, HMGA1a Protein metabolism, Multiple Myeloma pathology, Neoplasm Recurrence, Local pathology, RNA-Binding Proteins metabolism
- Abstract
Clonal evolution drives tumor progression, dissemination, and relapse in multiple myeloma (MM), with most patients dying of relapsed disease. This multistage process requires tumor cells to enter the circulation, extravasate, and colonize distant bone marrow (BM) sites. Here, we developed a fluorescent or DNA-barcode clone-tracking system on MM PrEDiCT (progression through evolution and dissemination of clonal tumor cells) xenograft mouse model to study clonal behavior within the BM microenvironment. We showed that only the few clones that successfully adapt to the BM microenvironment can enter the circulation and colonize distant BM sites. RNA sequencing of primary and distant-site MM tumor cells revealed a progression signature sequentially activated along human MM progression and significantly associated with overall survival when evaluated against patient data sets. A total of 28 genes were then computationally predicted to be master regulators (MRs) of MM progression. HMGA1 and PA2G4 were validated in vivo using CRISPR-Cas9 in the PrEDiCT model and were shown to be significantly depleted in distant BM sites, indicating their role in MM progression and dissemination. Loss of HMGA1 and PA2G4 also compromised the proliferation, migration, and adhesion abilities of MM cells in vitro. Overall, our model successfully recapitulates key characteristics of human MM disease progression and identified potential new therapeutic targets for MM., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
46. Can palliative care consultation increase integration of palliative care for patients with hematologic malignancies?
- Author
-
Prod'homme C, Touzet L, Pierrat M, Chevalier L, Lesaffre H, Berthon C, Coiteux V, Barbieux S, Beauvais D, Bauschert L, De Charette M, Goursaud L, Manier S, and Facon T
- Subjects
- Humans, Referral and Consultation, Hematologic Neoplasms therapy, Palliative Care
- Published
- 2021
- Full Text
- View/download PDF
47. Clonal hematopoiesis is associated with adverse outcomes in multiple myeloma patients undergoing transplant.
- Author
-
Mouhieddine TH, Sperling AS, Redd R, Park J, Leventhal M, Gibson CJ, Manier S, Nassar AH, Capelletti M, Huynh D, Bustoros M, Sklavenitis-Pistofidis R, Tahri S, Hornburg K, Dumke H, Itani MM, Boehner CJ, Liu CJ, AlDubayan SH, Reardon B, Van Allen EM, Keats JJ, Stewart C, Mehr S, Auclair D, Schlossman RL, Munshi NC, Anderson KC, Steensma DP, Laubach JP, Richardson PG, Ritz J, Ebert BL, Soiffer RJ, Trippa L, Getz G, Neuberg DS, and Ghobrial IM
- Subjects
- Adult, Aged, Aged, 80 and over, DNA (Cytosine-5-)-Methyltransferases genetics, DNA Methyltransferase 3A, DNA-Binding Proteins genetics, Dioxygenases, Female, Humans, Male, Middle Aged, Multiple Myeloma genetics, Progression-Free Survival, Proto-Oncogene Proteins genetics, Transplantation, Autologous, Tumor Suppressor Protein p53 genetics, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Hematopoiesis genetics, Hematopoietic Stem Cell Transplantation methods, Multiple Myeloma therapy, Mutation
- Abstract
Multiple myeloma (MM) is a plasma-cell neoplasm that is treated with high-dose chemotherapy, autologous stem cell transplant (ASCT) and long-term immunomodulatory drug (IMiD) maintenance. The presence of somatic mutations in the peripheral blood is termed clonal hematopoiesis of indeterminate potential (CHIP) and is associated with adverse outcomes. Targeted sequencing of the stem cell product from 629 MM patients treated by ASCT at the Dana-Farber Cancer Institute (2003-2011) detects CHIP in 136/629 patients (21.6%). The most commonly mutated genes are DNMT3A, TET2, TP53, ASXL1 and PPM1D. Twenty-one from fifty-six patients (3.3%) receiving first-line IMiD maintenance develop a therapy-related myeloid neoplasm (TMN). However, regardless of CHIP status, the use of IMiD maintenance associates with improved PFS and OS. In those not receiving IMiD maintenance, CHIP is associated with decreased overall survival (OS) (HR:1.34, p = 0.02) and progression free survival (PFS) (HR:1.45, p < 0.001) due to an increase in MM progression.
- Published
- 2020
- Full Text
- View/download PDF
48. Circulating tumor markers: harmonizing the yin and yang of CTCs and ctDNA for precision medicine.
- Author
-
Batth IS, Mitra A, Manier S, Ghobrial IM, Menter D, Kopetz S, and Li S
- Published
- 2019
- Full Text
- View/download PDF
49. Response to pneumococcal vaccination in multiple myeloma.
- Author
-
Renaud L, Schraen S, Fouquet G, Guidez S, Demarquette H, Nudel M, Cayssials E, Bories C, Herbaux C, Systchenko T, Faucompré JL, Machet A, Sabirou F, Levy A, Bobin A, Richez V, Moya N, Gruchet C, Desmier D, van de Wyngaert Z, Carpentier B, Manier S, Facon T, Harding S, and Leleu X
- Subjects
- Adult, Aged, Antibodies, Bacterial immunology, B-Lymphocytes immunology, B-Lymphocytes metabolism, Enzyme-Linked Immunosorbent Assay, Female, Humans, Immunologic Memory, Male, Middle Aged, Outcome Assessment, Health Care, Pneumococcal Vaccines administration & dosage, Vaccination, Multiple Myeloma complications, Pneumococcal Infections etiology, Pneumococcal Infections prevention & control, Pneumococcal Vaccines immunology
- Abstract
Background: Streptococcus pneumoniae infection causes morbidity and mortality in multiple myeloma patients. Pneumococcal vaccination is commonly given to immunocompromised myeloma patients; however response data are sparse. Here, we present longitudinal response data to pneumococcal vaccination in multiple myeloma patients., Method: Twenty-eight multiple myeloma patients were included, 25 of whom were newly diagnosed. All the patients received two vaccines Prevnar13® and Pneumo23®. Serotype-specific IgG was measured by ELISA for all 23 vaccine serotypes at baseline, and then sequentially at different time points postvaccination until treatment ended. Response to vaccination is available for 20 patients. The primary endpoint was the incidence rate of patients who obtained an isotype response serum concentration after vaccination. Secondary endpoints included detailed isotype increase, time to first increase, further assessment of a decreased anti-pneumococcal serum concentrations following treatment including autologous stem cell transplantation (ASCT), rate of infection with a special attention to pneumococcal infection., Results: The median age was 66 years and the male to female ratio was 0.6. Anti-pneumococcal capsular polysaccharide (anti-PCP23) IgG, IgG2, IgA, and IgM responses were detected within 1 week postvaccination. Response to at least one subtype of antibody was obtained in 85% (n = 17) of patients, for at least two subtypes in 65% (n = 13), for at least three subtypes in 55% (n = 11), and 2 patients responded to all four subtypes. The median increase in the concentration of anti-PCP23 isotypes was threefold following vaccination, with the highest increase observed when Pneumo23® was given more than 30 days after Prevnar13®. The anti-pneumococcal geometric mean concentration decreased significantly for all subtypes over time independently of treatment approaches., Conclusion: Myeloma has the ability to demonstrate a response to pneumococcal vaccine, independently of preexisting hypogammaglobulinemia and possibly of treatment-induced immunodepression. We also observed a drop in the serum response overtime and following autologous transplantation. Further studies in larger sample are needed to understand the benefit of vaccination strategies in these patients., (© 2019 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
50. Antigen excess pitfall for free light chains measurements solved by ELISA assay.
- Author
-
Pekar JD, Schraen S, Grzych G, Manier S, and Onraed B
- Subjects
- Enzyme-Linked Immunosorbent Assay, Humans, Male, Middle Aged, Antigens, Neoplasm blood, Immunoglobulin Light Chains blood, Multiple Myeloma blood, Neoplasm Proteins blood
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.