1,757 results on '"progression-free survival"'
Search Results
2. The CLL comorbidity index in a population-based cohort: a tool for clinical care and research
- Author
-
Emelie C. Rotbain, Max J. Gordon, Noomi Vainer, Henrik Frederiksen, Henrik Hjalgrim, Alexey V. Danilov, and Carsten U. Niemann
- Subjects
Cohort Studies ,immune system diseases ,hemic and lymphatic diseases ,Humans ,Comorbidity ,Hematology ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,neoplasms ,Progression-Free Survival - Abstract
The chronic lymphocytic leukemia comorbidity index (CLL-CI) is an efficient, CLL-specific tool derived from the Cumulative Illness Rating Scale. The CLL-CI is based on the assessment of the organ systems found to be most strongly associated with event-free survival (EFS) in CLL: vascular, upper gastrointestinal, and endocrine, at the time of initiation of CLL therapy. The CLL-CI categorizes patients into low, intermediate, and high risk groups. In the present study, we have employed the CLL-CI in a population-based cohort comprising 4975 patients with CLL. We demonstrate that CLL-CI retains prognostic significance in this large cohort and is associated with overall survival (OS) and EFS from time of first therapy. Furthermore, CLL-CI associates with OS, EFS, and time to first treatment from diagnosis independently of the CLL International Prognostic Index. These findings support the use of the CLL-CI both in research and in clinical practice.
- Published
- 2022
- Full Text
- View/download PDF
3. The impact of increasing karyotypic complexity and evolution on survival in patients with CLL treated with ibrutinib
- Author
-
Seema A. Bhat, Adam Kittai, Kyle A. Beckwith, Cecelia R. Miller, Ying Huang, Daniel Goldstein, Jennifer A. Woyach, Kerry A. Rogers, Lynne V. Abruzzo, Amy S. Ruppert, Michael R. Grever, John C. Byrd, David A. Bond, and Nyla A. Heerema
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,Immunology ,Abnormal Karyotype ,Disease ,Biochemistry ,Somatic evolution in cancer ,Clonal Evolution ,chemistry.chemical_compound ,Piperidines ,Refractory ,Chemoimmunotherapy ,Internal medicine ,medicine ,Humans ,Protein Kinase Inhibitors ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Venetoclax ,Adenine ,Cell Biology ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Survival Analysis ,Progression-Free Survival ,chemistry ,Ibrutinib ,Female ,business ,IGHV@ - Abstract
Complex karyotype, defined as ≥3 cytogenetic abnormalities, is prognostic of survival in patients treated with ibrutinib or venetoclax in relapsed/refractory (RR) chronic lymphocytic leukemia (CLL). Recent studies re-evaluating this dichotomous variable have shown that higher numbers of cytogenetic abnormalities (ie, ≥5) have a worse overall survival in patients treated with chemoimmunotherapy. We sought to determine if increasing karyotypic complexity, treated as a continuous variable, was prognostic of survival for patients treated with ibrutinib for CLL. We conducted a retrospective analysis of all patients with CLL treated with single-agent ibrutinib or in combination with an anti–CD20 antibody at our institution. We included 456 patients with both treatment-naive and RR disease. Median number of prior therapies was 2 (range, 0-13), 30% of patients had presence of del(17p), and 75% expressed unmutated IGHV. Fifty percent had ≥3 cytogenetic abnormalities, including 30% with ≥5. In a multivariable analysis, increasing karyotypic complexity was an independent predictor of shorter progression-free survival (hazard ratio, 1.07; 95% confidence interval, 1.04-1.10; P < .0001) and overall survival (hazard ratio, 1.09; 95% confidence interval, 1.05-1.12; P < .0001). Furthermore, we found that presence of clonal evolution determined by cytogenetic analysis at progression was prognostic of subsequent survival (P = .02). This solidifies karyotypic complexity as an important prognostic factor for patients with CLL treated with ibrutinib. Further research should consider sequential karyotypic analysis as a determination of risk of progression and death in patients with CLL.
- Published
- 2021
- Full Text
- View/download PDF
4. Impact of cryopreservation and transit times of allogeneic grafts on hematopoietic and immune reconstitution
- Author
-
Haesook T. Kim, John Koreth, Catherine J. Wu, Deborah Liney, Augustine Weber, Corey Cutler, Sarah Nikiforow, Katie Maurer, Jerome Ritz, Rizwan Romee, Mahasweta Gooptu, Roman M Shapiro, Joseph H. Antin, Vincent T. Ho, Robert J. Soiffer, Carol Reynolds, Thomas M. Kuczmarski, and Heather Garrity
- Subjects
Cryopreservation ,Oncology ,medicine.medical_specialty ,Platelet Engraftment ,SARS-CoV-2 ,business.industry ,T cell ,Hematopoietic Stem Cell Transplantation ,COVID-19 ,Context (language use) ,Hematology ,Allografts ,Haematopoiesis ,Immune Reconstitution ,medicine.anatomical_structure ,Immune system ,Internal medicine ,medicine ,Humans ,Progression-free survival ,Stem cell ,business ,Pandemics - Abstract
We sought to evaluate the impact of cryopreservation of unrelated donor (URD) peripheral blood stem cell (PBSC) grafts on engraftment, chimerism, and immune reconstitution in the context of the COVID-19 pandemic. We reviewed stem cell product characteristics and clinical outcomes in 101 patients receiving cryopreserved PBSCs from URDs between January 1, 2019 and 31 December, 2020, compared with 203 patients receiving fresh URD PBSCs. We observed no differences in 6-month overall survival, progression-free survival, or nonrelapse mortality. Patients receiving cryopreserved PBSCs had delayed platelet engraftment and impaired reconstitution of white blood cells and T-cell subsets at day 30. Thirty-four percent of patients receiving cryopreserved grafts had CD3 chimerism 48 hours old at time of cryopreservation or infusion significantly increased the risk of graft failure (subdistribution hazard ratio = 4.57; 95% confidence interval, 1.71-12.3; P = .0025). Our data indicate that cryopreservation is associated with similar overall short-term clinical outcomes compared with fresh PBSC. However, patients must be monitored closely for increased risk of other potentially adverse outcomes, including graft failure and poor immune recovery, particularly for grafts with older overall age at infusion. Longer-term follow-up is needed to determine impact on relapse and survival.
- Published
- 2021
- Full Text
- View/download PDF
5. Tisagenlecleucel immunogenicity in relapsed/refractory acute lymphoblastic leukemia and diffuse large B-cell lymphoma
- Author
-
Andy Warren, Michael A. Pulsipher, Theodore W. Laetsch, Stephen J. Schuster, G. Doug Myers, Peter Borchmann, John E. Levine, Michael Boyer, Edmund K. Waller, Edward Waldron, Stephan A. Grupp, Bernd Potthoff, Karen Thudium Mueller, Andrea Chassot-Agostinho, Ulrich Jaeger, Stephen Ronan Foley, Constantine S. Tam, Rakesh Awasthi, Keith J. August, Shannon L. Maude, and Fraser McBlane
- Subjects
medicine.medical_specialty ,Receptors, Antigen, T-Cell ,Cmax ,Gastroenterology ,Mice ,Immune system ,Refractory ,Internal medicine ,medicine ,Animals ,Humans ,Interferon gamma ,Child ,biology ,business.industry ,Immunogenicity ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Progression-Free Survival ,Lymphoma ,biology.protein ,Lymphoma, Large B-Cell, Diffuse ,Antibody ,business ,Diffuse large B-cell lymphoma ,medicine.drug - Abstract
Tisagenlecleucel is indicated for pediatric and young adult patients with relapsed/refractory (r/r) B-cell acute lymphoblastic leukemia (B-ALL) and adult patients with r/r diffuse large B-cell lymphoma (DLBCL). The tisagenlecleucel chimeric antigen receptor (CAR) contains a murine single-chain variable fragment domain; we examined the effects of humoral and cellular immune responses to tisagenlecleucel on clinical outcomes using 2 validated assays. Data were pooled from the ELIANA (registered at www.clinicaltrials.gov as #NCT02435849) and ENSIGN (#NCT02228096) trials in r/r B-ALL (N = 143) and the JULIET trial (#NCT02445248) in r/r DLBCL (N = 115). Humoral responses were determined by flow cytometric measurement of anti-murine CAR19 (mCAR19) antibodies in serum. Cellular responses were determined using T-cell production of interferon-γ in response to 2 different pools of mCAR19 peptides. Pretreatment anti-mCAR19 antibodies were detected in 81% of patients with r/r B-ALL and 94% of patients with r/r DLBCL. Posttreatment anti-mCAR19 antibodies were higher than patient-specific baseline in 42% of r/r B-ALL and 9% of r/r DLBCL patients. Pretreatment and posttreatment anti-mCAR19 antibodies did not affect tisagenlecleucel cellular kinetics, including maximum concentration and persistence (r2 < 0.05), clinical response (day-28 response, duration of response, and event-free survival), and safety. T-cell responses were consistent over time, with net responses
- Published
- 2021
- Full Text
- View/download PDF
6. CAR T-cell therapy for secondary CNS DLBCL
- Author
-
Mehdi Hamadani, Nirav N. Shah, and Gulrayz Ahmed
- Subjects
Oncology ,Central Nervous System ,medicine.medical_specialty ,Single Center ,Immunotherapy, Adoptive ,Refractory ,Internal medicine ,Medicine ,Humans ,Progression-free survival ,Complete response ,Retrospective Studies ,Receptors, Chimeric Antigen ,Adult patients ,business.industry ,Disease progression ,Hematology ,medicine.disease ,Stimulus Report ,Progression-Free Survival ,CAR T-cell therapy ,business ,Progressive disease - Abstract
Management of secondary central nervous system (SCNS) involvement in relapsed or refractory aggressive B-cell lymphomas remains an area of unmet medical need. We report a single-center retrospective analysis of 7 adult patients with SCNS lymphoma (SCNSL) who underwent chimeric antigen receptor (CAR) T-cell therapy for their refractory disease, and we describe the safety of whole brain radiation therapy (WBRT) as a bridging therapy. Six patients (85.7%) achieved a complete response at day 28, and 1 patient had progressive disease. The median progression-free survival was 83 days (range, 28-219 days), and median overall survival was 129 days (range, 32-219 days). Three patients died as a result of disease progression. Of the 5 patients who received WBRT as bridging therapy, 3 had no immune effector cell–associated neurotoxicity syndrome (ICANS), but 2 patients had grade 1 or grade 3 ICANS. No grade 4 ICANS was reported in this subset of patients. We conclude that SCNSL should not preclude patients from receiving CAR T-cell therapy as a treatment option because of concerns regarding ICANS, and bridging with WBRT is not associated with increased ICANS., Key Points SCNSL should not preclude patients from receiving CAR T-cell therapy because of concerns regarding ICANS.WBRT is not associated with increased ICANS when used as a bridge to CAR T-cell therapy with a short median interval in SCNSL.
- Published
- 2021
7. Outcomes of patients with blastoid and pleomorphic variant mantle cell lymphoma.
- Author
-
Gerson JN, Handorf E, Villa D, Gerrie AS, Chapani P, Li S, Medeiros LJ, Wang M, Cohen JB, Churnetski M, Hill BT, Sawalha Y, Hernandez-Ilizaliturri FJ, Kothari S, Vose JM, Bast M, Fenske T, Rao Gari SN, Maddocks KJ, Bond D, Bachanova V, Kolla B, Chavez J, Shah B, Lansigan F, Burns T, Donovan AM, Wagner-Johnston N, Messmer M, Mehta A, Anderson JK, Reddy N, Kovach AE, Landsburg DJ, Glenn M, Inwards DJ, Ristow K, Karmali R, Kaplan JB, Caimi PF, Rajguru S, Evens A, Klein A, Umyarova E, Pulluri B, Amengual JE, Lue JK, Diefenbach C, Fisher RI, and Barta SK
- Subjects
- Adult, Humans, Retrospective Studies, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Risk Assessment, Progression-Free Survival, Lymphoma, Mantle-Cell therapy, Lymphoma, Mantle-Cell drug therapy
- Abstract
Mantle cell lymphoma (MCL) is a B-cell non-Hodgkin lymphoma; data indicate that blastoid and pleomorphic variants have a poor prognosis. We report characteristics and outcomes of patients with blastoid/pleomorphic variants of MCL. We retrospectively studied adults with newly diagnosed MCL treated from 2000 to 2015. Primary objectives were to describe progression-free survival (PFS) and overall survival (OS). Secondary objectives included characterization of patient characteristics and treatments. Of the 1029 patients with MCL studied, a total of 207 neoplasms were blastoid or pleomorphic variants. Median follow-up period was 82 months (range, 0.1-174 months); median PFS was 38 months (95% confidence interval [CI], 28-66) and OS was 68 months (95% CI, 45-96). Factors associated with PFS were receipt of consolidative autologous hematopoietic transplantation (auto-HCT; hazard ratio [HR], 0.52; 95% CI, 0.31-0.80; P < .05), MCL International Prognostic Index (MIPI) intermediate (HR, 2.3; 95% CI, 1.2-4.3; P < .02) and high (HR, 3.8; 95% CI, 2.0-7.4; P < .01) scores, and complete response to induction (HR, 0.29 (95% CI, 0.17-0.51). Receipt of auto-HCT was not associated with OS (HR, 0.69; 95% CI, 0.41-1.16; P = .16) but was associated with MIPI intermediate (HR, 5.7; 95% CI, 2.5-13.2; P < .01) and high (HR, 10.8; 95% CI, 4.7-24.9; P < .01) scores. We report outcomes in a large cohort of patients with blastoid/pleomorphic variant MCL. For eligible patients, receipt of auto-HCT after induction was associated with improved PFS but not OS. Higher MIPI score and auto-HCT ineligibility were associated with worse survival., (© 2023 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
- 2023
- Full Text
- View/download PDF
8. High PDL1/PDL2 gene expression correlates with worse outcome in primary mediastinal large B-cell lymphoma.
- Author
-
Camus V, Viailly PJ, Drieux F, Veresezan EL, Sesques P, Haioun C, Durot E, Patey M, Rossi C, Martin L, Rainville V, Bohers E, Ruminy P, Penther D, Kaltenbach S, Bruneau J, Paillassa J, Tournilhac O, Willaume A, Antier C, Lazarovici J, Lévêque E, Decazes P, Becker S, Tonnelet D, Berriolo-Riedinger A, Gaulard P, Tilly H, Molina TJ, Traverse-Glehen A, and Jardin F
- Subjects
- Adolescent, Adult, Aged, Female, Humans, Middle Aged, Young Adult, Gene Expression, Prognosis, Progression-Free Survival, Proportional Hazards Models, Male, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Large B-Cell, Diffuse genetics
- Abstract
Primary mediastinal B-cell lymphoma (PMBL) is an uncommon entity of aggressive B-cell lymphoma with an unusually good prognosis, except for 10-15% of chemotherapy-refractory cases. To identify earlier these higher risk patients, we performed molecular characterization of a retrospective multicenter cohort of patients treated with firstline immunochemotherapy. The traits of the patients with gene-expression profiling data (n = 120) were as follows: median age of 34 years (range, 18-67 years); female sex, 58.3%; elevated lactate dehydrogenase, 82.5%; Eastern Cooperative Oncology Group performance status score of 0 to 1, 85.7%; Ann Arbor stage I/II, 55%; International Prognostic Index score of 1 to 2, 64.4%; and median metabolic tumor volume, 290.4 cm3 (range, 15.7-1147.5 cm3). Among all 137 markers tested for correlation with survival data, only programmed death-ligand (PDL) 1 and PDL2 expression showed a prognostic impact. Overall, both PDL1 and PDL2 genes were highly expressed in 37 patients (30.8%; PDL1high/PDL2high). The baseline clinical characteristics of patients with PDL1high/PDL2high were similar to those of other patients. In univariate analysis, PDL1high/PDL2high status was associated with poor progression-free survival (PFS) (hazard ratio [HR], 4.292) and overall survival (OS; HR, 8.24). In multivariate analysis, PDL1high/PDL2high status was an independent prognostic factor of adverse outcomes (PFS: HR, 5.22; OS: HR, 10.368). We validated these results in an independent cohort of 40 patients and confirmed the significant association between PDL1high/PDL2high status and inferior PFS (HR, 6.11). High PDL1/PDL2 gene expression defines a population with strong immune privilege and poorer outcomes from standard chemotherapy who might benefit from firstline checkpoint inhibitor therapy., (© 2023 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
- 2023
- Full Text
- View/download PDF
9. Prognostic model using 18F-FDG PET radiomics predicts progression-free survival in relapsed/refractory Hodgkin lymphoma.
- Author
-
Driessen J, Zwezerijnen GJC, Schöder H, Kersten MJ, Moskowitz AJ, Moskowitz CH, Eertink JJ, Heymans MW, Boellaard R, and Zijlstra JM
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Fluorodeoxyglucose F18, Positron-Emission Tomography methods, Prognosis, Progression-Free Survival, Transplantation, Autologous, Clinical Trials as Topic, Hematopoietic Stem Cell Transplantation, Hodgkin Disease therapy, Hodgkin Disease drug therapy
- Abstract
Investigating prognostic factors in patients with relapsed or primary refractory classical Hodgkin lymphoma (R/R cHL) is essential to optimize risk-adapted treatment strategies. We built a prognostic model using baseline quantitative 18F-fluorodeoxyglucose positron emission tomography (PET) radiomics features and clinical characteristics to predict the progression-free survival (PFS) among patients with R/R cHL treated with salvage chemotherapy followed by autologous stem cell transplantation. Metabolic tumor volume and several novel radiomics dissemination features, representing interlesional differences in distance, volume, and standard uptake value, were extracted from the baseline PET. Machine learning using backward selection and logistic regression were applied to develop and train the model on a total of 113 patients from 2 clinical trials. The model was validated on an independent external cohort of 69 patients. In addition, we validated 4 different PET segmentation methods to calculate radiomics features. We identified a subset of patients at high risk for progression with significant inferior 3-year PFS outcomes of 38.1% vs 88.4% for patients in the low-risk group in the training cohort (P < .001) and 38.5% vs 75.0% in the validation cohort (P = .015), respectively. The overall survival was also significantly better in the low-risk group (P = .022 and P < .001). We provide a formula to calculate a risk score for individual patients based on the model. In conclusion, we developed a prognostic model for PFS combining radiomics and clinical features in a large cohort of patients with R/R cHL. This model calculates a PET-based risk profile and can be applied to develop risk-stratified treatment strategies for patients with R/R cHL. These trials were registered at www.clinicaltrials.gov as #NCT02280993, #NCT00255723, and #NCT01508312., (© 2023 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
- 2023
- Full Text
- View/download PDF
10. RNA-seq of newly diagnosed patients in the PADIMAC study leads to a bortezomib/lenalidomide decision signature.
- Author
-
Chapman, Michael A., Koh, Mickey, Willis, Fenella, Virchis, Andres E., Crowe, Josephine, Quinn, Michael F., Cook, Gordon, Pratt, Guy, Cook, Mark, Owen, Roger G., Sonneveld, Pieter, Keats, Jonathan J., Crawley, Charles R., Sive, Jonathan, Cavenagh, Jamie D., Oakervee, Heather, Ambrose, John, Herrero, Javier, Roddie, Claire, and Popat, Rakesh
- Subjects
- *
BORTEZOMIB , *MULTIPLE myeloma , *RNA sequencing , *NEAREST neighbor analysis (Statistics) , *PROGRESSION-free survival , *MACHINE learning , *DOXORUBICIN , *DEXAMETHASONE - Abstract
Improving outcomes in multiple myeloma will involve not only development of new therapies but also better use of existing treatments. We performed RNA sequencing on samples from newly diagnosed patients enrolled in the phase 2 PADIMAC (Bortezomib, Adriamycin, and Dexamethasone Therapy for Previously Untreated Patients with Multiple Myeloma: Impact of Minimal Residual Disease in Patients with Deferred ASCT) study. Using synthetic annealing and the large margin nearest neighbor algorithm, we developed and trained a 7-gene signature to predict treatment outcome. We tested the signature in independent cohorts treated with bortezomib- and lenalidomide-based therapies. The signature was capable of distinguishing which patients would respond better to which regimen. In the CoMMpass data set, patients who were treated correctly according to the signature had a better progression-free survival (median, 20.1 months vs not reached; hazard ratio [HR], 0.40; confidence interval [CI], 0.23-0.72; P 5 .0012) and overall survival (median, 30.7 months vs not reached; HR, 0.41; CI, 0.21-0.80; P 5 .0049) than those who were not. Indeed, the outcome for these correctly treated patients was noninferior to that for those treated with combined bortezomib, lenalidomide, and dexamethasone, arguably the standard of care in the United States but not widely available elsewhere. The small size of the signature will facilitate clinical translation, thus enabling more targeted drug regimens to be delivered in myeloma. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
11. Prognostic impact of t(16;21)(p11;q22) and t(16;21)(q24;q22) in pediatric AML: a retrospective study by the I-BFM Study Group.
- Author
-
Noort, Sanne, Zimmermann, Martin, Reinhardt, Dirk, Cuccuini, Wendy, Pigazzi, Martina, Smith, Jenny, Ries, Rhonda E., Alonzo, Todd A., Hirsch, Betsy, Daisuke Tomizawa, Locatelli, Franco, Gruber, Tanja A., Raimondi, Susana, Sonneveld, Edwin, Cheuk, Daniel K., Dworzak, Michael, Stary, Jan, Abrahamsson, Jonas, Arad-Cohen, Nira, and Czogala, Malgorzata
- Subjects
- *
ACUTE myeloid leukemia in children , *ACUTE myeloid leukemia , *DISEASE relapse , *PROGRESSION-free survival , *CYTOGENETICS , *CHIMERIC proteins , *PROGNOSIS - Abstract
To study the prognostic relevance of rare genetic aberrations in acute myeloid leukemia (AML), such as t(16;21), international collaboration is required. Two different types of t(16;21) translocations can be distinguished: t(16;21)(p11;q22), resulting in the FUS-ERG fusion gene; and t(16;21)(q24;q22), resulting in RUNX1-core binding factor (CBFA2T3). We collected data on clinical and biological characteristics of 54 pediatric AML cases with t(16;21) rearrangements from 14 international collaborative study groups participating in the international Berlin-Frankfurt-Münster (I-BFM) AML study group. The AML-BFM cohort diagnosed between 1997 and 2013 was used as a reference cohort. RUNX1-CBFA2T3 (n = 23) had significantly lower median white blood cell count (12.5 3 109/L, P = .03) compared with the reference cohort. FUS-ERG rearranged AML (n = 31) had no predominant French-American- British (FAB) type, whereas 76% of RUNX1-CBFA2T3 had an M1/M2 FAB type (M1, M2), significantly different from the reference cohort (P = .004). Four-year event-free survival (EFS) of patients with FUS-ERG was 7% (standard error [SE] = 5%), significantly lower compared with the reference cohort (51%, SE = 1%, P < .001). Four-year EFS of RUNX1-CBFA2T3 was 77% (SE = 8%, P = .06), significantly higher compared with the reference cohort. Cumulative incidence of relapse was 74% (SE = 8%) in FUS- ERG, 0% (SE = 0%) in RUNX1-CBFA2T3, compared with 32% (SE = 1%) in the reference cohort (P < .001). Multivariate analysis identified both FUS-ERG and RUNX1-CBFA2T3 as independent risk factors with hazard ratios of 1.9 (P < .0001 ) and 0.3 (P = .025), respectively. These results describe 2 clinically relevant distinct subtypes of pediatric AML. Similarly to other core-binding factor AMLs, patients with RUNX1-CBFA2T3 rearranged AML may benefit from stratification in the standard risk treatment, whereas patients with FUS-ERG rearranged AML should be considered high-risk. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
12. Single-agent ibrutinib in treatment-naïve and relapsed/refractory chronic lymphocytic leukemia: a 5-year experience.
- Author
-
O'Brien, Susan, Furman, Richard R., Coutre, Steven, Flinn, Ian W., Burger, Jan A., Blum, Kristie, Sharman, Jeff, Wierda, William, Jones, Jeffrey, Weiqiang Zhao, Heerema, Nyla A., Johnson, Amy J., Ying Luan, James, Danelle F., Chu, Alvina D., and Byrd, John C.
- Subjects
- *
CANCER chemotherapy , *CANCER patients , *LYMPHOCYTIC leukemia , *CANCER treatment , *PROGRESSION-free survival - Abstract
We previously reported durable responses and manageable safety of ibrutinib from a 3-year follow-up of treatment-naïve (TN) older patients (≥65 years of age) and relapsed/refractory (R/R) patients with chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL). We now report on long-term efficacy and safety with median follow-up of 5 years in this patient population with TN (N = 31) and R/R (N = 101) CLL/SLL. With the current 5-year follow-up, ibrutinib continues to yield a high overall response rate of 89%, with complete response rates increasing over time to 29% in TN patients and 10% in R/R patients. The median progression-free survival (PFS) was not reached in TN patients. The 5-year PFS rate was 92% in TN patients and 44% in R/R patients. Median PFS in R/R patients was 51 months; in those with del(11q), del(17p), and unmutated IGHV, it was 51, 26, and 43 months, respectively, demonstrating long-term efficacy of ibrutinib in some high-risk subgroups. Survival outcomes were less robust for R/R patients with del(17p) and those who received more prior therapies. The onset of grade ≥3 cytopenias, such as neutropenia and thrombocytopenia, decreased over time. Treatment--limiting adverse events were more frequent during the first year compared with subsequent periods. These results demonstrate sustained efficacy and acceptable tolerability of ibrutinib over an extended time, providing the longest experience for Bruton tyrosine kinase inhibitor treatment in patients with CLL/SLL. These trials were registered at www.clinicaltrials.gov as #NCT01105247 and #NCT01109069. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
13. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia.
- Author
-
Gökbuget, Nicola, Dombret, Hervé, Bonifacio, Massimiliano, Reichle, Albrecht, Graux, Carlos, Faul, Christoph, Diedrich, Helmut, Topp, Max S., Brüggemann, Monika, Horst, Heinz-August, Havelange, Violaine, Stieglmaier, Julia, Wessels, Hendrik, Haddad, Vincent, Benjamin, Jonathan E., Zugmaier, Gerhard, Nagorsen, Dirk, and Bargou, Ralf C.
- Subjects
- *
LYMPHOBLASTIC leukemia , *B cells , *DISEASE remission , *HEMATOPOIETIC stem cell transplantation , *PROGRESSION-free survival - Abstract
Approximately 30% to 50% of adults with acute lymphoblastic leukemia (ALL) in hematologic complete remission after multiagent therapy exhibit minimal residual disease (MRD) by reverse transcriptase-polymerase chain reaction or flow cytometry. MRD is the strongest predictor of relapse in ALL. In this open-label, single-arm study, adults with B-cell precursor ALL in hematologic complete remission with MRD (≥10-3) received blinatumomab 15 μg/m2 per day by continuous IV infusion for up to 4 cycles. Patients could undergo allogeneic hematopoietic stem-cell transplantation any time after cycle 1. The primary end point was complete MRD response status after 1 cycle of blinatumomab. One hundred sixteen patients received blinatumomab. Eighty-eight (78%) of 113 evaluable patients achieved a complete MRD response. In the subgroup of 110 patients with Ph-negative ALL in hematologic remission, the Kaplan-Meier estimate of relapse-free survival (RFS) at 18 months was 54%. Median overall survival (OS) was 36.5 months. In landmark analyses, complete MRD responders had longer RFS (23.6 vs 5.7 months; P = .002) and OS (38.9 vs 12.5 months; P = .002) compared with MRD nonresponders. Adverse events were consistent with previous studies of blinatumomab. Twelve (10%) and 3 patients (3%) had grade 3 or 4 neurologic events, respectively. Four patients (3%) had cytokine release syndrome grade 1, n = 2; grade 3, n = 2), all during cycle 1. After treatment with blinatumomab in a population of patients with MRD-positive B-cell precursor ALL, a majority achieved a complete MRD response, which was associated with significantly longer RFS and OS compared with MRD nonresponders. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
14. Prognostic miRNA classifier in early-stage mycosis fungoides: development and validation in a Danish nationwide study.
- Author
-
Lindahl, Lise M., Besenbacher, Søren, Rittig, Anne H., Celis, Pamela, Willerslev-Olsen, Andreas, Gjerdrum, Lise M. R., Krejsgaard, Thørbjorn, Johansen, Claus, Litman, Thomas, Woetmann, Anders, Odum, Niels, and Iversen, Lars
- Subjects
- *
MICRORNA , *MYCOSIS fungoides , *CUTANEOUS T-cell lymphoma , *DISEASE progression , *PROGNOSIS , *PROGRESSION-free survival - Abstract
Mycosis fungoides (MF) is the most frequent form of cutaneous T-cell lymphoma. The disease often takes an indolent course, but in approximately one-third of the patients, the disease progresses to an aggressive malignancy with a poor prognosis. At the time of diagnosis, it is impossible to predict which patients develop severe disease and are in need of aggressive treatment. Accordingly, we investigated the prognostic potential of microRNAs (miRNAs) at the time of diagnosis in MF. Using a quantitative reverse transcription polymerase chain reaction platform, we analyzed miRNA expression in diagnostic skin biopsies from 154 Danish patients with early-stage MF. The patients were subdivided into a discovery cohort (n = 82) and an independent validation cohort (n = 72). The miRNA classifier was built using a LASSO (least absolute shrinkage and selection operator) Cox regression to predict progression-free survival (PFS). We developed a 3-miRNA classifier, based on miR-106b-5p, miR-148a-3p, and miR-338-3p, which successfully separated patients into high-risk and low-risk groups of disease progression. PFS was significantly different between these groups in both the discovery cohort and the validation cohort. The classifier was stronger than existing clinical prognostic factors and remained a strong independent prognostic tool after stratification and adjustment for these factors. Importantly, patients in the high-risk group had a significantly reduced overall survival. The 3-miRNA classifier is an effective tool to predict disease progression of early-stage MF at the time of diagnosis. The classifier adds significant prognostic value to existing clinical prognostic factors and may facilitate more individualized treatment of these patients. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
15. Expression of TP53 is associated with the outcome of MCL independent of MIPI and Ki-67 in trials of the European MCL Network.
- Author
-
Aukema, Sietse M., Hoster, Eva, Rosenwald, Andreas, Canoni, Danielle, Delfau-Larue, Marie-Hé léne, Rymkiewicz, Grzegorz, Thorns, Christoph, Hartmann, Sylvia, Kluin-Nelemans, Hanneke, Hermine, Olivier, Dreyling, Martin, and Klapper, Wolfram
- Subjects
- *
P53 protein , *PROTEIN expression , *MANTLE cell lymphoma , *HEALTH outcome assessment , *PROGRESSION-free survival , *IMMUNOHISTOCHEMISTRY , *MULTIVARIATE analysis - Abstract
Currently, prediction of time to treatment failure (TTF) and overall survival (OS) in mantle cell lymphoma (MCL) is based on the clinical factors included in the Mantle Cell Lymphoma International Prognostic Index (MIPI), and proliferation is assessed by Ki67. However, TP53 and SOX11 immunohistochemistry might improve risk stratification. We performed SOX11 and TP53 immunohistochemistry on the so far largest published cohort of lymphoma specimens (n 5 365). All patients were treated in prospective trials of the European MCL Network. In multivariate analyses, including MIPI and Ki67, SOX11 expression was not associated with TTF, but patients with low SOX11 expression had shorter OS. On the contrary, high TP53 expression was a strong predictor of TTF and inferior OS compared with low TP53 expression in univariate and multivariate analyses adjusting for MIPI score and Ki-67 index (hazard ratio [HR], 2.0; P 5 .0054 for TTF, and HR, 2.1; P 5 .068 for OS). In particular, patients with high TP53 expression (>50% positive lymphoma cells) had a shorter TTF and poor OS independent of both MIPI score and Ki-67 index. Thus, TP53 immunohistochemistry is a suitable test for routine diagnostic practice to assess MCL prognosis. (Blood. 2018;131(4):417-420) [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
16. CD38 antibodies in multiple myeloma: back to the future.
- Author
-
van de Donk, Niels W. C. J., Richardson, Paul G., and Malavasi, Fabio
- Subjects
- *
MULTIPLE myeloma treatment , *CD38 antigen , *THERAPEUTIC use of monoclonal antibodies , *IMMUNOLOGICAL adjuvants , *PROTEASOME inhibitors , *PROGRESSION-free survival , *THERAPEUTICS - Abstract
CD38 is highly and uniformly expressed on multiple myeloma (MM) cells, and at relatively low levels on normal lymphoid and myeloid cells, and in some tissues of nonhematopoietic origin. CD38 is a transmembrane glycoprotein with ectoenzymatic activity, and also functions as a receptor and adhesion molecule. Altogether, this has triggered the development of several CD38 antibodies including daratumumab (fully human), isatuximab (chimeric), and MOR202 (fully human). CD38 antibodies have pleiotropic mechanisms of action including Fc-dependent immune-effector mechanisms, direct apoptotic activity, and immunomodulatory effects by the elimination of CD381 immune-suppressor cells. CD38-targeting antibodies are generally well tolerated and induce partial response or better in ~30% of heavily pretreated MM patients as monotherapy. Based on their distinct mechanisms of action, favorable toxicity profile, and single-agent activity, CD38 antibodies are attractive partners in combination regimens. Indeed, deep responses and prolonged progression-free survival can be achieved in relapsed/refractory MM patients when CD38 antibodies are combined with immunomodulatory agents or proteasome inhibitors. Infusion-related reactions, which typically occur during the first infusion, are the most frequent adverse events. Attention should also be paid to the interference of CD38 antibodies with certain laboratory assays, which may complicate response evaluation and blood compatibility testing. Several studies are currently examining the role of CD38-based therapies in newly diagnosed and highrisk smoldering MM. Furthermore, CD38 antibodies are currently also under investigation in other hematologic malignancies, including acute lymphoblastic leukemia, natural killer/T-cell lymphoma, and acute myeloid leukemia, as well as in solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
17. Phase 2 study of the safety and efficacy of umbralisib in patients with CLL who are intolerant to BTK or PI3Kδ inhibitor therapy
- Author
-
Alan P Skarbnik, Paul M. Barr, Danielle M. Brander, Hanna Weissbrot, Ian W. Flinn, Peter Sportelli, Suman Kambhampati, Patricia Y. Tsao, Jeffrey Pu, Lindsey E. Roeker, Dana Paskalis, Nicole Lamanna, Stephen J. Schuster, Anthony R. Mato, James A. Reeves, Frederick Lansigan, Bruce D. Cheson, Michael S. Weiss, Nicole LaRatta, Gustavo Fonseca, Hari P. Miskin, Issam Hamadeh, Colleen Dorsey, Andrea Sitlinger, Nilanjan Ghosh, John M. Pagel, Eline T. Luning Prak, Kanti R. Rai, Jakub Svoboda, and Jacqueline C. Barrientos
- Subjects
medicine.medical_specialty ,education.field_of_study ,business.industry ,Immunology ,Population ,Phases of clinical research ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Gastroenterology ,Rash ,Discontinuation ,hemic and lymphatic diseases ,Internal medicine ,Clinical endpoint ,medicine ,Progression-free survival ,Leukocytosis ,medicine.symptom ,education ,business - Abstract
Purpose Intolerance is the most common reason for kinase inhibitor (KI) discontinuation in CLL. Umbralisib a novel, highly selective PI3Kδ/CK1e inhibitor, is active and well tolerated in CLL patients. This phase 2 trial evaluated umbralisib in CLL patients who are intolerant to prior BTK or PI3K inhibitor therapy. Patients and methods In this phase 2 trial (NCT02742090), umbralisib was initiated at 800 mg oral daily in CLL patients requiring therapy per investigator discretion who were intolerant to prior BTK or PI3K inhibitor therapy, until progression or toxicity. Primary endpoint was progression-free survival (PFS). Secondary endpoints included time to treatment failure and umbralisib safety profile. DNA isolated from buccal swabs was genotyped for polymorphisms in CYP3A4, CYP3A5 and CYP2D6. Results Fifty-one patients were enrolled (44 BTKi and 7 PI3Kδi intolerant). Median age was 70 years (range 48-96), median of 2 prior lines of therapy (1-7), 24% had del17p and/or TP53 mutation, and 65% were IGHV unmutated. Most common AEs leading to prior KI discontinuation were rash (27%), arthralgia (18%), and atrial fibrillation (16%). Median progression free survival (PFS) was 23.5 months (95% CI 13.1-not estimable). 58% of patients were on umbralisib for a longer duration than prior KI. Most common (≥5%) grade ≥3 AEs on umbralisib (all causality) were neutropenia (18%), leukocytosis (14%), thrombocytopenia (12%), pneumonia (12%), and diarrhea (8%). Six patients (12%) discontinued umbralisib due to an AE. Eight patients (16%) had dose reductions and were successfully re-challenged. Conclusions Umbralisib is safe and effective in this BTK and alternate PI3K inhibitor intolerant CLL population. These are the first prospective data to confirm that switching from a BTK or alternate PI3K inhibitor to umbralisib can result in durable, well tolerated responses.
- Published
- 2021
- Full Text
- View/download PDF
18. COVID-19 and hematopoietic stem cell transplantation and immune effector cell therapy: a US cancer center experience
- Author
-
Julie Porter, Cindy Albert, Haesook T. Kim, Sarah Nikiforow, Corey Cutler, Mahasweta Gooptu, Clifton C. Mo, Katie Maurer, Rizwan Romee, Utkarsh Acharya, Joseph H. Antin, Caron A. Jacobson, John Koreth, Catherine J. Wu, Anna Saucier, Robert J. Soiffer, and Vincent T. Ho
- Subjects
Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Graft vs Host Disease ,Kaplan-Meier Estimate ,Disease ,Hematopoietic stem cell transplantation ,Immunotherapy, Adoptive ,Transplantation, Autologous ,Young Adult ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Progression-free survival ,Aged ,Transplantation ,Neutrophil Engraftment ,SARS-CoV-2 ,Donor selection ,business.industry ,Hematopoietic Stem Cell Transplantation ,COVID-19 ,Hematology ,Middle Aged ,medicine.disease ,Progression-Free Survival ,United States ,Chimeric antigen receptor ,Cytokine release syndrome ,surgical procedures, operative ,Female ,business - Abstract
Key Points With sufficient resources, HSCT can safely continue in the COVID-19 pandemic if primary responsibility for COVID-19 patients is not required. Cryopreservation of unrelated donor products correlated with slightly lower chimerism but no difference in clinical outcomes at 100 days., The novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), identified in late 2019 as the causative agent of COVID-19, was declared a pandemic by the World Health Organization on 11 March 2020. Widespread community transmission in the United States triggered a nationwide shutdown, raising major challenges for administration of hematopoietic stem cell transplant (HSCT) and chimeric antigen receptor (CAR)-T cell therapies, leading many centers to delay or cancel operations. We sought to assess the impact of the COVID-19 pandemic on operations and clinical outcomes for HSCT and CAR-T cellular therapies at the Dana-Farber Cancer Institute by reviewing administration and outcomes in 127 cell therapy patients treated during the initial COVID-19 surge: 62 adult allogeneic HSCT (allo-HSCT), 38 autologous HSCT (auto-HSCT), and 27 CAR-T patients. Outcomes were compared with 66 allo-HSCT, 43 auto-HSCT, and 33 CAR-T patients treated prior to the pandemic. A second control cohort was evaluated for HSCT groups to reflect seasonal variation in infections. Although there were changes in donor selection and screening as well as cryopreservation patterns of donor products, no differences were observed across groups in 100-day overall survival, progression-free survival, rates of non–COVID-19 infections, including hospital length of stay, neutrophil engraftment, graft failure, acute graft-versus-host disease in allo-HSCT patients, or cytokine release syndrome and neurotoxicity in CAR-T patients. No HSCT patients contracted COVID-19 between days 0 and 100. One CAR-T patient contracted COVID-19 at day +51 and died of the disease. Altogether, our data indicate that cellular therapies can be safely administered throughout the ongoing COVID-19 pandemic with appropriate safeguards., Visual Abstract
- Published
- 2021
- Full Text
- View/download PDF
19. Denosumab compared with zoledronic acid on PFS in multiple myeloma: exploratory results of an international phase 3 study
- Author
-
Xavier Leleu, Waltraud Pasteiner, Peter I. Croucher, Yang Wang, Evangelos Terpos, Charlotte Pawlyn, Anthony Glennane, Noopur Raje, Jude Canon, and Ramón García-Sanz
- Subjects
Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Phases of clinical research ,Renal function ,Transplantation, Autologous ,Zoledronic Acid ,Autologous stem-cell transplantation ,Internal medicine ,medicine ,Humans ,Multiple myeloma ,Aged ,Bortezomib ,business.industry ,Hazard ratio ,Hematopoietic Stem Cell Transplantation ,Hematology ,medicine.disease ,Progression-Free Survival ,Denosumab ,Zoledronic acid ,Multiple Myeloma ,business ,medicine.drug - Abstract
An exploratory end point from a recent trial in patients with newly diagnosed multiple myeloma showed that median progression-free survival (PFS) was increased by 10.7 months with denosumab vs zoledronic acid. We performed additional analyses to identify factors that may have contributed to the favorable PFS with denosumab. Ad hoc analyses were performed for patients intending to undergo autologous stem cell transplantation (ASCT; ASCT intent), not intending to undergo ASCT (ASCT no intent), and intent-to-treat according to age (60 mL/min creatinine clearance [CrCl]). Of 1718 patients, 930 (54.1%) were in the ASCT-intent subgroup, and 788 (45.9%) were in the ASCT-no-intent subgroup. In the ASCT-intent subgroup, frontline triplet (median PFS, not estimable vs 35.7 months; hazard ratio [HR] [95% confidence interval (CI)], 0.65 [0.47-0.90]; descriptive P = .009) or bortezomib-only (median PFS, not estimable vs not estimable; HR [95% CI], 0.61 [0.39–0.95]; descriptive P = .029) induction regimens demonstrated the strongest PFS benefit favoring denosumab vs zoledronic acid. In the ASCT-no-intent subgroup, no benefit with denosumab vs zoledronic acid was observed. PFS favored denosumab vs zoledronic acid in patients with CrCl >60 mL/min and in patients
- Published
- 2021
- Full Text
- View/download PDF
20. Double unrelated umbilical cord blood vs HLA-haploidentical bone marrow transplantation: the BMT CTN 1101 trial
- Author
-
Richard J. Jones, Andrew R. Rezvani, Lawrence E. Morris, Peter Dawson, Joseph P. McGuirk, Eric S. Leifer, John M. Magenau, Paul O'Donnell, Chatchada Karanes, Steven M. Devine, Ephraim J. Fuchs, Mary M. Horowitz, Mary Eapen, Joseph H. Antin, Claudio G. Brunstein, Mitchell E. Horwitz, and Brent R. Logan
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Cyclophosphamide ,Immunology ,Graft vs Host Disease ,Biochemistry ,Umbilical cord ,Gastroenterology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,HLA Antigens ,Cause of Death ,Internal medicine ,medicine ,Humans ,Progression-free survival ,Aged ,Bone Marrow Transplantation ,Acute leukemia ,business.industry ,Incidence ,Cell Biology ,Hematology ,Middle Aged ,Total body irradiation ,Fetal Blood ,Progression-Free Survival ,Hematopoiesis ,Transplantation ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cord blood ,Acute Disease ,Chronic Disease ,Transplantation, Haploidentical ,Female ,Bone marrow ,Unrelated Donors ,business ,medicine.drug - Abstract
Results of two parallel phase II trials of transplantation of unrelated umbilical cord blood or bone marrow from HLA-haploidentical relatives provided equipoise for direct comparison of these donor sources. Between June 2012 and June 2018, 368 patients aged 18-70 years with chemotherapy-sensitive lymphoma or acute leukemia in remission were randomly assigned to undergo cord blood (n=186) or haploidentical (n=182) transplant. Reduced intensity conditioning comprised total body irradiation with cyclophosphamide and fludarabine for both donor types. Graft-versus-host disease prophylaxis for cord blood transplantation was cyclosporine and mycophenolate mofetil and for haploidentical transplantation, post-transplant cyclophosphamide, tacrolimus and mycophenolate mofetil. The primary endpoint was 2-year progression-free survival. Treatment groups had similar age, sex, self-reported ethnic origin, performance status, disease and disease status at randomization. Two-year progression-free survival was 35% (95% CI, 28-42%) compared to 41% (95% CI, 34-48%) after cord blood and haploidentical transplants, respectively (p=0.41). Pre-specified analysis of secondary endpoints recorded higher 2-year non-relapse mortality after cord blood, 18% (95% CI, 13-24%) compared to haploidentical transplantation, 11% (95% CI, 6-16%), p=0.04. This led to lower 2-year overall survival after cord blood compared to haploidentical transplantation, 46% (95% CI, 38-53) and 57% (95% CI 49-64%), respectively (p=0.04). The trial did not demonstrate a statistically significant difference in the primary endpoint, 2-year progression-free survival, between the donor sources. While both donor sources extend access to reduced intensity transplantation, analyses of secondary endpoints, including overall survival, favor haploidentical bone marrow donors. (Funded by the National Heart, Lung, and Blood Institute-National Cancer Institute; ClinicalTrials.gov number, NCT01597778).
- Published
- 2021
- Full Text
- View/download PDF
21. Active surveillance of primary extranodal marginal zone lymphoma of bronchus-associated lymphoid tissue
- Author
-
Esther Drill, Steven M. Horwitz, Carol S. Portlock, Sridevi Rajeeve, Matthew J. Matasar, Anas Younes, Ariela Noy, John F. Gerecitano, Erel Joffe, Yan Leyfman, Craig H. Moskowitz, David J. Straus, Connie Lee Batlevi, Anita Kumar, M. Lia Palomba, Andrew D. Zelenetz, Alison J. Moskowitz, and Paul A. Hamlin
- Subjects
medicine.medical_specialty ,Lymphoid Neoplasia ,Lung ,business.industry ,medicine.medical_treatment ,Cancer ,Bronchi ,Retrospective cohort study ,Lymphoma, B-Cell, Marginal Zone ,Hematology ,Disease ,Immunotherapy ,medicine.disease ,Gastroenterology ,Progression-Free Survival ,Lymphoma ,Lymphatic system ,medicine.anatomical_structure ,Internal medicine ,medicine ,Humans ,Lymph ,Watchful Waiting ,business ,Retrospective Studies - Abstract
Although patients with bronchus-associated lymphoid tissue (BALT) lymphoma show an indolent clinical course, appropriate disease management at diagnosis is not well defined. This study aimed to compare 3 treatment strategies for patients with BALT lymphoma: active surveillance, systemic chemotherapy or immunotherapy at diagnosis, or complete surgical resection at diagnosis. We conducted a retrospective study of all patients with new diagnoses of marginal zone lymphoma (MZL) involving the lung who were treated at the Memorial Sloan Kettering Cancer Center between 1995 and 2017. Primary BALT lymphoma was defined as disease confined to the lungs and adjacent lymph nodes. Active surveillance was defined as a documented observation plan and ≥3 months of follow-up before initiating treatment. Overall survival (OS) and event-free survival (EFS) were compared between treatment groups. We reviewed 200 consecutive patients with MZL involving the lung; 123 met the inclusion criteria and were managed by active surveillance (47%), complete surgical resection (41%), or systemic chemotherapy or immunotherapy (11%). With a median follow-up of >60 months, surgical resection was associated with a superior EFS compared with active surveillance and systemic treatment (6-year EFS: 74% vs 65% vs 62%, respectively; P = .013). Larger lesions and thrombocytopenia were associated with shorter EFS. All groups had excellent OS at 6 years (93%), albeit with a slight superiority for surgical resection (100%) over active surveillance (91%) and systemic treatment (76%) (P = .024). BALT lymphoma is an indolent disease that can often be managed expectantly and not require therapy for many years.
- Published
- 2021
- Full Text
- View/download PDF
22. Ixazomib significantly prolongs progression-free survival in high-risk relapsed/refractory myeloma patients.
- Author
-
Avet-Loiseau, Hervé, Bahlis, Nizar J., Wee-Joo Chng, Masszi, Tamas, Viterbo, Luisa, Pour, Ludek, Ganly, Peter, Palumbo, Antonio, Cavo, Michele, Langer, Christian, Pluta, Andrzej, Nagler, Arnon, Kumar, Shaji, Ben-Yehuda, Dina, Rajkumar, S. Vincent, San-Miguel, Jesus, Berg, Deborah, Jianchang Lin, van de Velde, Helgi, and Esseltine, Dixie-Lee
- Subjects
- *
MULTIPLE myeloma treatment , *PROTEASOME inhibitors , *PROGRESSION-free survival , *DEXAMETHASONE , *CYTOGENETICS , *THERAPEUTICS - Abstract
Certain cytogenetic abnormalities are known to adversely impact outcomes in patients with multiple myeloma (MM). The phase 3 TOURMALINE-MM1 study demonstrated a significant improvement in progression-free survival (PFS) with ixazomib-lenalidomide-dexamethasone (IRd) compared with placebo-lenalidomide-dexamethasone (placebo-Rd). This preplanned analysis assessed the efficacy and safety of IRd vs placebo-Rd according to cytogenetic risk, as assessed using fluorescence in situ hybridization. High-risk cytogenetic abnormalities were defined as del(17p), t(4;14), and/or t(14;16); additionally, patients were assessed for 1q21 amplification. Of 722 randomized patients, 552 had cytogenetic results; 137 (25%) had high-risk cytogenetic abnormalities and 172 (32%) had 1q21 amplification alone. PFS was improved with IRd vs placebo-Rd in both high-risk and standard-risk cytogenetics subgroups: in high-risk patients, the hazard ratio (HR) was 0.543 (95% confidence interval [CI], 0.321-0.918; P = .021), with median PFS of 21.4 vs 9.7 months; in standard-risk patients, HR was 0.640 (95%CI, 0.462- 0.888; P = .007), with median PFS of 20.6 vs 15.6 months. This PFS benefit was consistent across subgroups with individual high-risk cytogenetic abnormalities, including patients with del(17p) (HR, 0.596; 95% CI, 0.286-1.243). PFS was also longer with IRd vs placebo-Rd in patients with 1q21 amplification (HR, 0.781; 95% CI, 0.492-1.240), and in the "expanded high-risk" group, defined as those with high-risk cytogenetic abnormalities and/or 1q21 amplification (HR, 0.664; 95% CI, 0.474-0.928). IRd demonstrated substantial benefit compared with placebo-Rd in relapsed and/or refractory MM (RRMM) patients with high-risk and standard-risk cytogenetics, and improves the poor PFS associated with high-risk cytogenetic abnormalities. This trial was registered at www.clinicaltrials.gov as #NCT01564537. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. Clonal chromosomal abnormalities appearing in Philadelphia chromosome-negative metaphases during CML treatment.
- Author
-
Issa, Ghayas C., Kantarjian, Hagop M., Gonzalez, Graciela Nogueras, Borthakur, Gautam, Guilin Tang, Wierda, William, Koji Sasaki, Short, Nicholas J., Ravandi, Farhad, Kadia, Tapan, Patel, Keyur, Luthra, Raja, Ferrajoli, Alessandra, Garcia-Manero, Guillermo, Rios, Mary Beth, Dellasala, Sara, Jabbour, Elias, and Cortes, Jorge E.
- Subjects
- *
TREATMENT of chronic myeloid leukemia , *CHROMOSOME abnormalities , *METAPHASE (Mitosis) , *PROTEIN-tyrosine kinase inhibitors , *TRISOMY , *PROGRESSION-free survival - Abstract
Clonal chromosomal abnormalities in Philadelphia chromosome-negative metaphases (CCA/Ph-) emerge as patients with chronic phase chronic myeloid leukemia (CP-CML) are treated with tyrosine kinase inhibitors (TKI). We assessed the characteristics and prognostic impact of 598 patients with CP-CML treated on clinical trials with various TKIs. CCA/Ph- occurred in 58 patients (10%) and the most common were - Y in 25 (43%) and trisomy 8 in 7 patients (12%). Response to TKI therapy was similar for patients with CCA/Ph- and those without additional chromosomal abnormalities (ACAs). We further categorized CCA/Ph- into those where - Y was the only clonal abnormality, and all others. We found that patients with non -Y CCA/Ph- had worse failure-free survival (FFS), event-free survival (EFS), transformation-free survival (TFS) and overall survival (OS) compared to those without ACAs with the following 5-year rates: FFS (52% vs 70%, P=0.02), EFS (68% vs 86%, P=0.02), TFS (76% vs 94%, P <0.01) and OS (79% vs 94%, P=0.03). In a multivariate analysis, non -Y CCA/Ph- increased the risk of transformation or death when baseline characteristics were considered with a hazard ratio (HR) of 2.81 (95% CI 1.15-6.89, P=0.02). However, this prognostic impact was not statistically significant when achieving BCR-ABL <10% at 3 months was included in the analysis. In conclusion, non -Y CCA/Ph- are associated with decreased survival when emerging in patients with chronic phase CML across various TKIs. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
24. Daratumumab plus pomalidomide and dexamethasone in relapsed and/or refractory multiple myeloma.
- Author
-
Chari, Ajai, Suvannasankha, Attaya, Fay, Joseph W., Arnulf, Bertrand, Kaufman, Jonathan L., Ifthikharuddin, Jainulabdeen J., Weiss, Brendan M., Krishnan, Amrita, Lentzsch, Suzanne, Comenzo, Raymond, Jianping Wang, Nottage, Kerri, Chiu, Christopher, Khokhar, Nushmia Z., Ahmadi, Tahamtan, and Lonial, Sagar
- Subjects
- *
MULTIPLE myeloma treatment , *THALIDOMIDE , *THERAPEUTIC use of monoclonal antibodies , *DEXAMETHASONE , *DISEASE relapse , *NEUTROPENIA , *COMBINATION drug therapy , *PROGRESSION-free survival , *THERAPEUTICS - Abstract
Daratumumab plus pomalidomide and dexamethasone (pom-dex) was evaluated in patients with relapsed/refractory multiple myeloma with ≥2 prior lines of therapy who were refractory to their last treatment. Patients received daratumumab 16 mg/kg at the recommended dosing schedule, pomalidomide 4 mg daily for 21 days of each 28-day cycle, and dexamethasone 40 mg weekly. Safety was the primary end point. Overall response rate (ORR) and minimal residual disease (MRD) by next-generation sequencing were secondary end points. Patients (N 5 103) received a median (range) of 4 (1-13) prior therapies; 76% received ≥3 prior therapies. The safety profile of daratumumab plus pomdex was similar to that of pom-dex alone, with the exception of daratumumab-specific infusion-related reactions (50%) and a higher incidence of neutropenia, although without an increase in infection rate. Common grade ≥3 adverse events were neutropenia (78%), anemia (28%), and leukopenia (24%). ORR was 60% and was generally consistent across subgroups (58% in double-refractory patients). Amongpatients with a complete response or better, 29% were MRD negative at a threshold of 1025. Among the 62 responders, median duration of response was not estimable (NE; 95% confidence interval [CI], 13.6-NE). At a median follow-up of 13.1 months, the median progression-free survival was 8.8 (95% CI, 4.6-15.4) months andmedian overall survivalwas17.5 (95% CI, 13.3-NE) months. The estimated 12-month survival ratewas 66% (95% CI, 55.6-74.8). Aside from increased neutropenia, the safety profile of daratumumab plus pom-dex was consistent with that of the individual therapies. Deep, durable responses were observed in heavily treated patients. The study was registered at www.clinicaltrials.gov as #NCT01998971. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
25. How I treat first relapse of myeloma.
- Author
-
Harousseau, Jean Luc and Attal, Michel
- Subjects
- *
MULTIPLE myeloma treatment , *DEXAMETHASONE , *BORTEZOMIB , *DISEASE relapse , *PROGRESSION-free survival , *THALIDOMIDE , *DRUG resistance in cancer cells , *THERAPEUTICS - Abstract
The standard treatment of relapsed multiple myeloma has been either lenalidomide-dexamethasone (RD) or bortezomib-dexamethasone (VD) but it is changing rapidly for 2 reasons. First, lenalidomide and bortezomib are currently used in frontline treatment and many patients become resistant to these agents early in the course of their disease. Second, 6 second-line newagents have beenrecently developed and offer new possibilities (pomalidomide, carfilzomib and ixazomib, panobinostat, elotuzumab, and daratumumab). Recent randomized studies have shown that triple combinations adding 1 of these new agents (except pomalidomide) to the RD or VD regimens were superior to the double combinations in terms of response rate and progressionfree survival (PFS). Their place in the treatment of first relapse is discussed here. Among these agents, daratumumab is clearly a breakthrough and daratumumabbased combinations might become the preferred option in the near future. However, all of these drugs are expensive and are not available or affordable in all countries. We propose a decision algorithm for first relapse in fit patients with the objective of achieving the best PFS. The choice of salvage regimen is based on lenalidomide/bortezomib resistance, daratumumabavailability, andcost. Autologous transplantation should be considered in youngerpatientsif notusedupfront. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
26. A phase 1b study of isatuximab plus lenalidomide and dexamethasone for relapsed/refractory multiple myeloma.
- Author
-
Martin, Thomas, Baz, Rachid, Benson, Don M., Lendvai, Nikoletta, Wolf, Jeffrey, Munster, Pamela, Lesokhin, Alexander M., Wack, Claudine, Charpentier, Eric, Campana, Frank, and Vij, Ravi
- Subjects
- *
MYELOID leukemia , *LEUKEMIA treatment , *DOWN syndrome , *DEXAMETHASONE , *PROGRESSION-free survival , *FLOW cytometry , *PATIENTS - Abstract
Patients with myeloid leukemia of Down syndrome (ML-DS) have favorable event-free survival (EFS), but experience significant treatment-related morbidity and mortality. ML-DS blast cells ex vivo have increased sensitivity to cytarabine (araC) and daunorubicin, suggesting that optimizing drug dosing may improve outcomes while reducing toxicity. The Children's Oncology Group (COG) AAML0431 trial consisted of 4 cycles of induction and 2 cycles of intensification therapy based on the treatment schema of the previous COG A2971 trial with several modifications. High-dose araC (HD-araC) was used in the second induction cycle instead of the intensification cycle, and 1 of 4 daunorubicin-containing induction cycles was eliminated. For 204 eligible patients, 5-year EFS was 89.9% and overall survival (OS) was 93.0%. The 5-year OS for 17 patients with refractory/relapsed leukemia was 34.3%. We determined the clinical significance of minimal residual disease (MRD) levels as measured by flow cytometry on day 28 of induction I. MRD measurements, available for 146 of the 204 patients, were highly predictive of treatment outcome; 5-year disease-free survival for MRD-negative patients (n = 125) was 92.7% vs 76.2% for MRD-positive patients (n = 21) (log-rank P = .011). Our results indicated that earlier use of HD-araC led to better EFS and OS in AAML0431 than in past COG studies. A 25% reduction in the cumulative daunorubicin dose did not impact outcome. MRD, identified as a new prognostic factor for ML-DS patients, can be used for risk stratification in future clinical trials. This trial was registered at www.clinicaltrials.gov as #NCT00369317. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
27. Rituximab exposure is influenced by baseline metabolic tumor volume and predicts outcome of DLBCL patients: a Lymphoma Study Association report.
- Author
-
Tout, Mira, Casasnovas, Olivier, Meignan, Michel, Lamy, Thierry, Morschhauser, Franck, Salles, Gilles, Gyan, Emmanuel, Haioun, Corinne, Mercier, Mélanie, Feugier, Pierre, Boussetta, Sami, Paintaud, Gilles, Ternant, David, and Cartron, Guillaume
- Subjects
- *
DIFFUSE large B-cell lymphomas , *RITUXIMAB , *CANCER chemotherapy , *PROGRESSION-free survival , *HEALTH outcome assessment , *COMPUTED tomography , *PHARMACOKINETICS , *THERAPEUTICS - Abstract
High variability in patient outcome after rituximab-based treatment is partly explained by rituximab concentrations, and pharmacokinetic (PK) variability could be influenced by tumor burden. We aimed at quantifying the influence of baseline total metabolic tumor volume (TMTV0) on rituximab PK and of TMTV0 and rituximab exposure on outcome in patients with diffuse large B-cell lymphoma (DLBCL). TMTV0 was measured by 18F-fluorodeoxyglucosepositron emission tomography-computed tomography in 108 previously untreated DLBCL patients who received four 375mg/m² rituximab infusions every 2 weeks in combinationwith chemotherapy in 2 prospective trials. A 2-compartment populationmodel allowed describing rituximab PK and calculating rituximab exposure (area under the concentration-time curve; AUC). The association of TMTV0 and AUC with metabolic response after 4 cycles, as well as progression-free survival (PFS) and overall survival (OS), was assessed using logistic regression and Coxmodels, respectively. Cutoff values for patient outcomewere determined using receiver operating characteristic curve analysis. Exposure to rituximab decreased as TMTV0 increased (R² 5 0.41, P < .0001). A high AUC in cycle 1 (≥ 9400 mg x h per liter) was associated withbetter response (oddsratio, 5.56;P5.0006) and longerPFS(hazard ratio [HR], 0.38;P5.011) and OS(HR, 0.17; P = 5.001). A nomogram for rituximab dose needed to obtain optimal AUC according to TMTV0 was constructed, and the 375 mg/m² classical dose would be suitable for patients with TMTV0 <281 cm³. In summary, rituximab exposure is influenced by TMTV0 and correlates with response and outcome of DLBCL patients. Dose individualization according to TMTV0 should be evaluated in prospective studies. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
28. Radiation-free, alternative-donor HCT for Fanconi anemia patients: results from a prospective multi-institutional study.
- Author
-
Mehta, Parinda A., Davies, Stella M., Leemhuis, Thomas, Myers, Kasiani, Kernan, Nancy A., Prockop, Susan E., Scaradavou, Andromachi, O'Reilly, Richard J., Williams, David A., Lehmann, Leslie, Guinan, Eva, Margolis, David, Baker, K. Scott, Lane, Adam, and Boulad, Farid
- Subjects
- *
HEMATOPOIETIC stem cell transplantation , *FANCONI'S anemia , *GRAFT versus host disease , *RADIATION exposure , *BUSULFAN , *CYCLOPHOSPHAMIDE , *PROGRESSION-free survival , *PATIENTS - Abstract
Fanconi anemia (FA) is an inherited bone marrow failure syndrome characterized by chromosomal fragility, progressive marrow failure, and cancer predisposition. Hematopoietic cell transplantation (HCT) is curative for FA-related marrow failure or leukemia, but both radiation exposure during transplant and graft-versus-host disease (GVHD) may increase risk of later malignancies of the head and neck and anogenital area. In this study, we tested a radiation-free conditioning regimen with a T-cell--depleted graft to eliminate radiation exposure and minimize early and late toxicities of transplant. Forty-five patients (median age, 8.2 years; range 4.3-44) with FA underwent HCT between June 2009 and May 2014. The preparative regimen included busulfan, cyclophosphamide, fludarabine, and rabbit anti-thymocyte globulin. Busulfan levels were monitored to avoid excess toxicity. All grafts were CD34-selected/T-cell--depleted using the CliniMacs CD34 columns (Miltenyi). Thirty-four patients (75.6%) with marrow failure and 11 (24.4%) with myelodysplastic syndrome underwent HCT using matched unrelated (n = 25, 55.5%), mismatched unrelated (n = 14, 31.1%), or mismatched related donors (n = 6, 13.4%). One year probabilities of overall and disease-free survival for the entire cohort, including patients with myeloid malignancy and those receiving mismatched related/haploidentical grafts, were 80% (±6%) and 77.7% (±6.2%), respectively (median follow-up 41 months). All young children (<10 years of age) undergoing HCT for marrow failure using low-dose busulfan-containing regimen survived. No patients developed acute grade 3-4 GVHD. Sequential reduction of busulfan dose was successfully achieved per study design. Our results show excellent outcomes in patients with FA undergoing alternative donor HCT without radiation exposure. The study is registered to www.clinicaltrials.gov as #NCT01082133. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
29. Evaluating surveillance imaging for diffuse large B-cell lymphoma and Hodgkin lymphoma.
- Author
-
Cohen, Jonathon B., Behera, Madhusmita, Thompson, Carrie A., and Flowers, Christopher R.
- Subjects
- *
BCL-2 proteins , *HODGKIN'S disease , *DISEASE relapse , *PROGRESSION-free survival , *CLINICAL competence , *PATIENTS - Abstract
Up to 50% of patients with Hodgkin lymphoma and diffuse large B-cell lymphoma will relapse, requiring additional therapy. Although surveillance imaging is commonly performed in clinical practice, its ability to identify asymptomatic relapses and improve survival for patients is not well defined. We evaluated the surveillance imaging role in relapse detection and reviewed its impact on survival for relapsed patients, and found that current imaging approaches do not detect most relapses prior to clinical signs and symptoms or improve survival. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
30. Mutations associated with progression in follicular lymphoma predict inferior outcomes at diagnosis: Alliance A151303.
- Author
-
Russler-Germain DA, Krysiak K, Ramirez C, Mosior M, Watkins MP, Gomez F, Skidmore ZL, Trani L, Gao F, Geyer S, Cashen AF, Mehta-Shah N, Kahl BS, Bartlett NL, Alderuccio JP, Lossos IS, Ondrejka SL, Hsi ED, Martin P, Leonard JP, Griffith M, Griffith OL, and Fehniger TA
- Subjects
- Humans, Risk Factors, Prognosis, Progression-Free Survival, Mutation, Lymphoma, Follicular diagnosis, Lymphoma, Follicular genetics
- Abstract
Follicular lymphoma (FL) is clinically heterogeneous, with select patients tolerating extended watch-and-wait, whereas others require prompt treatment, suffer progression of disease within 24 months of treatment (POD24), and/or experience aggressive histologic transformation (t-FL). Because our understanding of the relationship between genetic alterations in FL and patient outcomes remains limited, we conducted a clinicogenomic analysis of 370 patients with FL or t-FL (from Cancer and Leukemia Group B/Alliance trials 50402/50701/50803, or real-world cohorts from Washington University School of Medicine, Cleveland Clinic, or University of Miami). FL subsets by grade, stage, watch-and-wait, or POD24 status did not differ by mutation burden, whereas mutation burden was significantly higher in relapsed/refractory (rel/ref) FL and t-FL than in newly diagnosed (dx) FL. Nonetheless, mutation burden in dx FL was not associated with frontline progression-free survival (PFS). CREBBP was the only gene more commonly mutated in FL than in t-FL yet mutated CREBBP was associated with shorter frontline PFS in FL. Mutations in 20 genes were more common in rel/ref FL or t-FL than in dx FL, including 6 significantly mutated genes (SMGs): STAT6, TP53, IGLL5, B2M, SOCS1, and MYD88. We defined a mutations associated with progression (MAP) signature as ≥2 mutations in these 7 genes (6 rel/ref FL or t-FL SMGs plus CREBBP). Patients with dx FL possessing a MAP signature had shorter frontline PFS, revealing a 7-gene set offering insight into FL progression risk potentially more generalizable than the m7-Follicular Lymphoma International Prognostic Index (m7-FLIPI), which had modest prognostic value in our cohort. Future studies are warranted to validate the poor prognosis associated with a MAP signature in dx FL, potentially facilitating novel trials specifically in this high-risk subset of patients., (© 2023 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
- 2023
- Full Text
- View/download PDF
31. Low toxicity and excellent outcomes in patients with DLBCL without residual lymphoma at the time of CD19 CAR T-cell therapy.
- Author
-
Wudhikarn K, Tomas AA, Flynn JR, Devlin SM, Brower J, Bachanova V, Nastoupil LJ, McGuirk JP, Maziarz RT, Oluwole OO, Schuster SJ, Porter DL, Bishop MR, Riedell PA, and Perales MA
- Subjects
- Humans, Immunotherapy, Adoptive adverse effects, Progression-Free Survival, Remission Induction, Antigens, CD19, Lymphoma, Large B-Cell, Diffuse drug therapy, Lymphoma, Non-Hodgkin drug therapy
- Abstract
CD19 chimeric antigen receptor (CAR) T-cell therapy represents a breakthrough for patients with relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL), inducing sustained remissions in these patients. However, CAR T cells can result in significant toxicities. Preinfusion disease burden is associated with toxicities and outcomes after CAR T-cell therapy. We identified 33 patients with R/R DLBCL treated at 8 academic centers who had no detectable disease at the time of CAR T-cell therapy. The median time from leukapheresis to CAR T-cell infusion was 48 (19-193) days. Nine patients received axicabtagene ciloleucel, and 24 received tisagenlecleucel. There was no severe (grade ≥3) cytokine release syndrome, and only 1 patient developed severe neurotoxicity (grade 4). After a median follow-up of 16 months, 13 patients relapsed (39.4%) and 6 died (18.1%). One-year event-free survival and overall survival were 59.6% and 81.3%, respectively. Our findings suggest that, in patients with R/R DLBCL who have an indication for CAR T-cell therapy, treating patients in complete remission at the time of infusion is feasible, safe, and associated with favorable disease control. Further exploration in a larger clinical trial setting is warranted., (© 2023 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
- 2023
- Full Text
- View/download PDF
32. Lenalidomide-associated B-cell ALL: clinical and pathologic correlates and sensitivity to lenalidomide withdrawal.
- Author
-
Geyer MB, Shaffer BC, Bhatnagar B, Mims AS, Klein V, Dilip D, Glass JL, Lozanski G, Hassoun H, Landau H, Zhang Y, Xiao W, Roshal M, and Park JH
- Subjects
- Humans, Aged, Lenalidomide, Retrospective Studies, Progression-Free Survival, Multiple Myeloma therapy, Hematopoietic Stem Cell Transplantation adverse effects, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Burkitt Lymphoma drug therapy
- Abstract
Lenalidomide is an effective component of induction and maintenance therapy for multiple myeloma, though with a risk of secondary malignancies, including acute lymphoblastic leukemia (ALL). In contrast to therapy-related myeloid neoplasia, lenalidomide-associated lymphoblastic neoplasia remains poorly characterized. We conducted a dual institution retrospective study of 32 ALL cases that arose after lenalidomide maintenance (all B-lineage, 31/32 BCR::ABL-negative). B-cell ALL (B-ALL) was diagnosed at median 54 months (range, 5-119) after first exposure to lenalidomide and after median 42 months of cumulative lenalidomide exposure (range, 2-114). High incidence of TP53 mutations (9/19 evaluable cases) and low hypodiploidy (8/26 patients) were identified. Despite median age of 65 years and poor-risk B-ALL features observed in the cohort, rates of complete response (CR) or CR with incomplete hematologic recovery were high (25/28 patients receiving treatment). Median event-free survival was 35.4 months among treated patients (not reached among those undergoing allogeneic hematopoietic cell transplantation [HCT]). Sixteen patients remain alive without evidence of B-ALL after HCT or extended maintenance therapy. We also describe regression of B-ALL or immature B-cell populations with B-ALL immunophenotype after lenalidomide discontinuation in 5 patients, suggesting lenalidomide may drive leukemic progression even after initiation of lymphoblastic neoplasia and that lenalidomide withdrawal alone may be an appropriate first-line intervention in selected patients. Monitoring for early B-ALL-like proliferations may offer opportunities for lenalidomide withdrawal to prevent progression. Established combination chemotherapy regimens, newer surface antigen-targeted approaches, and allogeneic HCT are effective in many patients with lenalidomide-associated B-ALL and should be offered to medically fit patients., (© 2023 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
- 2023
- Full Text
- View/download PDF
33. Carfilzomib, lenalidomide, and dexamethasone plus transplant in newly diagnosed multiple myeloma
- Author
-
Ravi Vij, Andrzej Jakubowiak, Kathryn M. Tinari, Kent A. Griffith, Leonor A Stephens, Donna E. Reece, Sarah Major, Andrew T. Stefka, Alexandra E Rojek, Brittany Wolfe, Tyler Hycner, Sandeep Gurbuxani, Jagoda Jasielec, Jesus G. Berdeja, Benjamin A. Derman, Todd M. Zimmerman, Tadeusz Kubicki, Cara A. Rosenbaum, Paul G. Richardson, Dominik Dytfeld, and Noopur Raje
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Clinical Trials and Observations ,Immunology ,Neutropenia ,Biochemistry ,Dexamethasone ,Disease-Free Survival ,Autologous stem-cell transplantation ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Progression-free survival ,Autografts ,Lenalidomide ,Multiple myeloma ,Aged ,business.industry ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Minimal residual disease ,Progression-Free Survival ,Regimen ,Tolerability ,Female ,Multiple Myeloma ,business ,Oligopeptides ,medicine.drug - Abstract
In this phase 2 multicenter study, we evaluated the incorporation of autologous stem cell transplantation (ASCT) into a carfilzomib-lenalidomide-dexamethasone (KRd) regimen for patients with newly diagnosed multiple myeloma (NDMM). Transplant-eligible patients with NDMM received 4 cycles of KRd induction, ASCT, 4 cycles of KRd consolidation, and 10 cycles of KRd maintenance. The primary end point was rate of stringent complete response (sCR) after 8 cycles of KRd with a predefined threshold of ≥50% to support further study. Seventy-six patients were enrolled with a median age of 59 years (range, 40-76 years), and 35.5% had high-risk cytogenetics. The primary end point was met, with an sCR rate of 60% after 8 cycles. Depth of response improved over time. On intent-to-treat (ITT), the sCR rate reached 76%. The rate of minimal residual disease (MRD) negativity using modified ITT was 70% according to next-generation sequencing (
- Published
- 2020
- Full Text
- View/download PDF
34. Predictive factors of early progression after CAR T-cell therapy in relapsed/refractory diffuse large B-cell lymphoma
- Author
-
Maud D'Aveni-Piney, Catherine Thieblemont, Caroline Bodet-Milin, Loic Ysebaert, Pierre Feugier, Eugenio Galli, Marie-Thérèse Rubio, Roberta Di Blasi, Pierre Bories, Benoit Tessoulin, Pierre Olivier, Ingrid Lafon, Alina Berriolo-Riedinger, Sylvie Chevret, Sophie Bernard, Laetitia Vercellino, Céline Bossard, Salim Kanoun, Lucie Oberic, Pascal Merlet, Michel Meignan, Olivier Casasnovas, Steven Le Gouill, Véronique Meignin, Cédric Rossi, Hopital Saint-Louis [AP-HP] (AP-HP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Service de Médecine Nucléaire [Toulouse], CHU Toulouse [Toulouse], Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM), Service d'Hématologie [CHRU Nancy], Centre Hospitalier Régional Universitaire de Nancy (CHRU Nancy), Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Université de Lorraine (UL)-Centre National de la Recherche Scientifique (CNRS), Service d'Hématologie [IUCT Toulouse], Université Fédérale Toulouse Midi-Pyrénées-Institut Universitaire du Cancer de Toulouse - Oncopole (IUCT Oncopole - UMR 1037), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-CHU Toulouse [Toulouse]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-CHU Toulouse [Toulouse]-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de Médecine Nucléaire [Nantes], Hôpital Laennec, Service de Médecine Nucléaire [Nancy], Service de Médecine Nucléaire, Centre Georges-François Leclerc [Dijon] (CGFL), Centre Régional de Lutte contre le cancer Georges-François Leclerc [Dijon] (UNICANCER/CRLCC-CGFL), UNICANCER-UNICANCER, Centre hospitalier universitaire de Nantes (CHU Nantes), Nutrition-Génétique et Exposition aux Risques Environnementaux (NGERE), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lorraine (UL), CHU Henri Mondor, Service de biostatistiques et information médicale [Saint-Louis], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Groupe Hospitalier Saint Louis - Lariboisière - Fernand Widal [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université de Paris (UP), NF-kappaB, Différenciation et Cancer (OncokappaB (URP_7324)), and Université de Paris (UP)
- Subjects
0301 basic medicine ,medicine.medical_specialty ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Immunotherapy, Adoptive ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,International Prognostic Index ,Refractory ,Internal medicine ,medicine ,Humans ,Progression-free survival ,Stage (cooking) ,Retrospective Studies ,Univariate analysis ,business.industry ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Hematology ,medicine.disease ,Progression-Free Survival ,Confidence interval ,Tumor Burden ,3. Good health ,Lymphoma ,030104 developmental biology ,030220 oncology & carcinogenesis ,Lymphoma, Large B-Cell, Diffuse ,business ,Diffuse large B-cell lymphoma - Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as an option for relapsed/refractory aggressive B-cell lymphomas that have failed 2 lines of therapy. Failures usually occur early after infusion. The purpose of our study was to identify factors that may predict failure, particularly early progression (EP), within the first month after infusion. Characteristics of 116 patients were analyzed at the time of decision (TD) to use commercial CAR (axicabtagene ciloleucel, n = 49; tisagenlecleucel n = 67) and at the time of treatment (TT), together with total metabolic tumor volume (TMTV) at TT. With a median follow-up of 8.2 months, 55 patients failed treatment; 27 (49%) were early progressors. The estimated 12-month progression-free survival (PFS) and overall survival (OS) were 47.2% (95% confidence interval [CI], 38.0-58.6) and 67.0% (95% CI, 57-79), respectively. Univariate analyses for PFS and OS identified Eastern Cooperative Oncology Group Performance Status (ECOG PS) ≥2, stage III/IV disease, extranodal (EN) sites ≥2, elevated lactate dehydrogenase (LDH), increased C-reactive protein (CRP), high International Prognostic Index at TD and at TT, as well as increased CRP, bulky mass, and high TMTV at TT, as risk factors. Multivariate analyses for PFS, EP, and OS identified elevated LDH and EN sites ≥2 at TD and the same predictors at TT (ie, increased CRP, EN sites ≥2, and TMTV >80 mL). In summary, risk factors identified for early progression at TD and at TT were EN involvement (≥2 sites) and lymphoma burden (LDH, TMTV).
- Published
- 2020
- Full Text
- View/download PDF
35. Risk score to predict event-free survival after hematopoietic cell transplant for sickle cell disease
- Author
-
Mary Eapen, Ruta Brazauskas, Damiano Rondelli, Barbara Cappelli, Courtney D. Fitzhugh, Julie Kanter, Hai-Lin Wang, Jane S. Hankins, Julie A. Panepinto, John E. Wagner, Shalini Shenoy, Mark C. Walters, Eliane Gluckman, Graziana Maria Scigliuolo, John F. Tisdale, Joerg J Meerpohl, and Annalisa Ruggeri
- Subjects
Adult ,Male ,medicine.medical_specialty ,Allogeneic transplantation ,Adolescent ,Immunology ,Anemia, Sickle Cell ,Biochemistry ,Young Adult ,Risk Factors ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Progression-free survival ,Young adult ,Child ,Transplantation ,Framingham Risk Score ,Proportional hazards model ,business.industry ,Histocompatibility Antigens Class I ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Sickle cell anemia ,Treatment Outcome ,Blood Grouping and Crossmatching ,Child, Preschool ,Population study ,Female ,business - Abstract
We developed a risk score to predict event-free survival (EFS) after allogeneic hematopoietic cell transplantation for sickle cell disease. The study population (n = 1425) was randomly split into training (n = 1070) and validation (n = 355) cohorts. Risk factors were identified and validated via Cox regression models. Two risk factors of 9 evaluated were predictive for EFS: age at transplantation and donor type. On the basis of the training cohort, patients age 12 years or younger with an HLA-matched sibling donor were at the lowest risk with a 3-year EFS of 92% (score, 0). Patients age 13 years or older with an HLA-matched sibling donor or age 12 years or younger with an HLA-matched unrelated donor were at intermediate risk (3-year EFS, 87%; score, 1). All other groups, including patients of any age with a haploidentical relative or HLA-mismatched unrelated donor and patients age 13 years or older with an HLA-matched unrelated donor were high risk (3-year EFS, 57%; score, 2 or 3). These findings were confirmed in the validation cohort. This simple risk score may guide patients with sickle cell disease and hematologists who are considering allogeneic transplantation as a curative treatment relative to other available contemporary treatments.
- Published
- 2020
- Full Text
- View/download PDF
36. Clinical and biological implications of target occupancy in CLL treated with the BTK inhibitor acalabrutinib
- Author
-
Sarah E. M. Herman, Hailey Harris, Priti Patel, Raquel Izumi, Maryalice Stetler-Stevenson, Constance M. Yuan, Michael Gulrajani, Todd Covey, Irina Maric, Christopher Pleyer, Jean Cheung, Adrian Wiestner, Stefania Pittaluga, Min Hui Wang, Clare Sun, Pia Nierman, Mohammed Farooqui, Ellen K. Kendall, Erika M Gaglione, Inhye E. Ahn, and Ahmed Hamdy
- Subjects
Male ,Chronic lymphocytic leukemia ,Immunology ,Pain ,Phases of clinical research ,Antineoplastic Agents ,Pharmacology ,Biochemistry ,Drug Administration Schedule ,immune system diseases ,hemic and lymphatic diseases ,Agammaglobulinaemia Tyrosine Kinase ,Humans ,Medicine ,Bruton's tyrosine kinase ,RNA, Messenger ,RNA, Neoplasm ,RNA-Seq ,Dosing ,Progression-free survival ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,biology ,business.industry ,Headache ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Hematologic Diseases ,Leukemia, Lymphocytic, Chronic, B-Cell ,Progression-Free Survival ,Neoplasm Proteins ,Leukemia ,Treatment Outcome ,Enzyme Induction ,Pyrazines ,Pharmacodynamics ,Benzamides ,biology.protein ,Acalabrutinib ,Female ,Transcriptome ,business - Abstract
Inhibition of the B-cell receptor pathway, and specifically of Bruton tyrosine kinase (BTK), is a leading therapeutic strategy in B-cell malignancies, including chronic lymphocytic leukemia (CLL). Target occupancy is a measure of covalent binding to BTK and has been applied as a pharmacodynamic parameter in clinical studies of BTK inhibitors. However, the kinetics of de novo BTK synthesis, which determines occupancy, and the relationship between occupancy, pathway inhibition and clinical outcomes remain undefined. This randomized phase 2 study investigated the safety, efficacy, and pharmacodynamics of a selective BTK inhibitor acalabrutinib at 100 mg twice daily (BID) or 200 mg once daily (QD) in 48 patients with relapsed/refractory or high-risk treatment-naïve CLL. Acalabrutinib was well tolerated and yielded an overall response rate (ORR) of partial response or better of 95.8% (95% confidence interval [CI], 78.9-99.9) and an estimated progression-free survival (PFS) rate at 24 months of 91.5% (95% CI, 70.0-97.8) with BID dosing and an ORR of 79.2% (95% CI, 57.9-92.9) and an estimated PFS rate at 24 months of 87.2% (95% CI, 57.2-96.7) with QD dosing. BTK resynthesis was faster in patients with CLL than in healthy volunteers. BID dosing maintained higher BTK occupancy and achieved more potent pathway inhibition compared with QD dosing. Small increments in occupancy attained by BID dosing relative to QD dosing compounded over time to augment downstream biological effects. The impact of BTK occupancy on long-term clinical outcomes remains to be determined. This trial was registered at www.clinicaltrials.gov as #NCT02337829.
- Published
- 2020
- Full Text
- View/download PDF
37. Prognostic and predictive impact of genetic markers in patients with CLL treated with obinutuzumab and venetoclax
- Author
-
Stephan Stilgenbauer, Michele Porro Lurà, Karl-Anton Kreuzer, Yanwen Jiang, Barbara Eichhorst, Eugen Tausch, Michael Hallek, Sandra Robrecht, Jasmin Bahlo, Can Zhang, Hartmut Döhner, Christof Schneider, Johannes Bloehdorn, Anna-Maria Fink, William Schary, Daniel Mertens, Maneesh Tandon, Kirsten Fischer, Anna Dolnik, Kathryn Humphrey, Othman Al-Sawaf, Matthias Ritgen, Lars Bullinger, and Michael Kneba
- Subjects
Genetic Markers ,Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,Immunology ,Immunoglobulin Variable Region ,Kaplan-Meier Estimate ,Gene mutation ,Antibodies, Monoclonal, Humanized ,Biochemistry ,chemistry.chemical_compound ,Chemoimmunotherapy ,Obinutuzumab ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Multicenter Studies as Topic ,Progression-free survival ,Chromosome Aberrations ,Sulfonamides ,Chlorambucil ,business.industry ,Venetoclax ,Remission Induction ,Hazard ratio ,Cell Biology ,Hematology ,Bridged Bicyclo Compounds, Heterocyclic ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,Progression-Free Survival ,Clinical Trials, Phase III as Topic ,chemistry ,Mutation ,Immunoglobulin Heavy Chains ,IGHV@ ,business ,Follow-Up Studies ,Genes, Neoplasm ,medicine.drug - Abstract
Genetic parameters are established prognostic factors in chronic lymphocytic leukemia (CLL) treated with chemoimmunotherapy, but are less well studied with novel compounds. We assessed immunoglobulin heavy variable chain (IGHV) mutation status, common genomic aberrations, and gene mutations in 421 untreated patients within the CLL14 trial (NCT02242942), comparing obinutuzumab+chlorambucil (GClb) vs obinutuzumab+venetoclax (VenG). The incidences of genomic aberrations considering the hierarchical model were del(17p) 7%, del(11q) 18%, +12 18%, and del(13q) 35%, whereas IGHV was unmutated in 60% of patients. NOTCH1 mutations were most common (23%), followed by SF3B1 (16%), ATM (13%), and TP53 (10%). Although the overall response rate (ORR) for GClb was lower in patients with del(17p), del(11q), mutated TP53, ATM, and BIRC3, none of these parameters reduced complete remission (CR) rate and ORR with VenG. At a median follow-up of 28 months, del(17p) and mutated TP53 were the only abnormalities with an effect on progression-free survival (PFS) for both treatment groups: GClb (hazard ratio [HR], 4.6 [P < .01]; HR, 2.7 [P < .01], respectively) and VenG (HR, 4.4 [P < .01]; HR, 3.1 [P < .01], respectively). No other factors affected outcome with VenG, whereas for GClb del(11q), BIRC3, NOTCH1, and unmutated IGHV were associated with shorter PFS. Multivariable analysis identified del(17p), del(11q), unmutated IGHV, and mutated TP53, BIRC3, and SF3B1 as independent prognostic factors for PFS with GClb, whereas for VenG, only del(17p) was significant. VenG was superior to GClb across most genetic subgroups. Patients with adverse genetic markers had the strongest benefit from VenG, particularly subjects with unmutated IGHV, which was identified as a predictive factor in a multivariable treatment-interaction analysis.
- Published
- 2020
- Full Text
- View/download PDF
38. Clinical value of event-free survival in acute myeloid leukemia
- Author
-
Naval Daver, Courtney D. DiNardo, Gautam Borthakur, Hagop M. Kantarjian, Helen O. Ajufo, Guillermo Garcia-Manero, Jorge E. Cortes, William G. Wierda, Srdan Verstovsek, Marina Konopleva, Michael Andreeff, Rohit V. Goswamy, Tapan M. Kadia, Vinita Popat, Farhad Ravandi, Steven M. Kornblau, Miguel Velasquez, Carlos Blanco, Naveen Pemmaraju, Abhishek Maiti, and Sherry Pierce
- Subjects
medicine.medical_specialty ,Myeloid Neoplasia ,Blood transfusion ,business.industry ,medicine.medical_treatment ,Remission Induction ,Hematopoietic Stem Cell Transplantation ,MEDLINE ,Myeloid leukemia ,Retrospective cohort study ,Hematology ,Chemotherapy regimen ,Progression-Free Survival ,Transplantation ,Leukemia, Myeloid, Acute ,Blood product ,Internal medicine ,Health care ,Humans ,Medicine ,business ,Retrospective Studies - Abstract
The value of event-free survival (EFS) as an end point in acute myeloid leukemia (AML) trials has been questioned. We hypothesized that rather than a surrogate for overall survival (OS), improvement in EFS may decrease the use of health care. In this retrospective study, we identified 400 patients with AML who were treated on first-line therapy trials and had OS between 2 and 36 months. We captured health care use from diagnosis until death or until the patient was censored at stem cell transplantation (SCT). We used correlation and regression analysis to determine the relation between health care use and EFS. Among patients with newly diagnosed AML, 35% had adverse-risk AML, 48% received intensive chemotherapy, and 28% received hypomethylating agents. The median EFS censored at SCT was 9.7 months. Longer EFS led to a significant decline in health care use regardless of OS. This held true for all observations, including overall health care use (r = −0.45), sum of clinic visits, emergency room visits, hospitalizations, consultations (r = −0.44), sum of invasive procedures, laboratory and imaging studies (r = −0.51), and blood product transfusions (r = −0.19). These correlations were stronger for patients who achieved a complete remission and held true across age, treatment, and disease risk subgroups. In patients with newly diagnosed AML, improvement in EFS correlates with a decrease in all health care use irrespective of OS duration.
- Published
- 2020
- Full Text
- View/download PDF
39. Maximum tumor diameter is associated with event-free survival in PET-negative patients with stage I/IIA Hodgkin lymphoma
- Author
-
Elizabeth H Phillips, Andrew McMillan, Ruth Pettengell, Peter Hoskin, Nicholas Counsell, Sally F. Barrington, John Radford, Dominic Culligan, Laura Clifton-Hadley, Timothy M Illidge, Peter Johnson, and Bilyana Popova
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Progression-free survival ,Manchester Cancer Research Centre ,medicine.diagnostic_test ,Tumor size ,business.industry ,ResearchInstitutes_Networks_Beacons/mcrc ,Event free survival ,Hematology ,Hodgkin Disease ,Stimulus Report ,Progression-Free Survival ,Radiation therapy ,030104 developmental biology ,ABVD ,Positron emission tomography ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Hodgkin lymphoma ,business ,medicine.drug - Abstract
Introduction: the high cure rates achieved in early-stage (ES) Hodgkin lymphoma (HL) are one of the great successes of hemato-oncology, but late treatment-related toxicity undermines long-term survival. Improving overall survival and quality of life further will require maintaining disease control while potentially de-escalating chemotherapy and/or omitting radiotherapy to reduce late toxicity. Accurate stratification of patients is required to facilitate individualized treatment approaches. Response assessment using 18F-fluorodeoxyglucose positron emission tomography (PET) is a powerful predictor of outcome in HL,1,2 and has been used in multiple studies, including the United Kingdom National Cancer Research Institute Randomised Phase III Trial to Determine the Role of FDG–PET Imaging in Clinical Stages IA/IIA Hodgkin’s Disease (UK NCRI RAPID) trial, to investigate whether patients achieving complete metabolic remission (CMR) can be treated with chemotherapy alone.3-5 These PET-adapted trials have demonstrated that omitting radiotherapy results in higher relapse rates, but without compromising overall survival.3-5 For the 75% of patients who achieved CMR in RAPID, neither baseline clinical risk stratification (favorable/unfavorable) nor PET (Deauville score 1/2) predicted disease relapse; additional biomarkers are needed.1 Tumor bulk has long been recognized as prognostic in HL,1,6 but there remains uncertainty about the significance and definition of bulk in the era of PET-adapted treatment.7 We performed a subsidiary analysis of RAPID to assess the prognostic value of baseline maximum tumor dimension (MTD) in patients achieving CMR. Methods: ee have previously reported the RAPID trial design, primary results, and outcomes according to pretreatment risk stratification and PET score.1,3 Patients were aged 16 to 75 years with untreated ES-HL and without B-symptoms or mediastinal bulk (mass > 1/3 internal mediastinal diameter at T5/6).6 Metabolic response after 3 cycles of ABVD chemotherapy (doxorubicin, bleomycin, vinblastine, and dacarbazine) was centrally assessed using PET (N = 562). Patients with CMR (ie, Deauville score 1-2) were randomly assigned to receive involved field radiotherapy (IFRT; n = 208) or no further therapy (NFT; n = 211). PET-positive patients (score, 3-5; n = 143) received a fourth cycle of ABVD and IFRT. Baseline disease assessment was performed by computed tomography, and bidimensional target lesion measurements were reported by local radiologists in millimeters. The association of baseline MTD with HL-related event-free survival (EFS: progression or HL-related death) and progression-free survival (PFS) (progression or any-cause death) was assessed using Kaplan-Meier and Cox regression analyses. Non-HL deaths were either related to primary treatment toxicity or occurred in HL remission.1 United Kingdom ethical approval for the RAPID trial was via the UK Multicentre Research ethics committee. Results and discussion: baseline patient characteristics have been previously described.1 Median age was 34 years (range, 16-75 years); 184 (37.4%) of 492 patients had unfavorable risk by European Organisation for Research and Treatment of Cancer criteria, and 155 (32.3%) of 480 by German Hodgkin Study Groupcriteria. Median MTD for patients achieving CMR was 3.0 cm (interquartile range, 2.0-4.0 cm) and 3.0 cm (interquartile range, 1.8-4.5 cm) in the NFT and IFRT groups, respectively, whereas PET-positive patients had a median MTD of 3.9 cm (interquartile range, 2.8-5.1 cm). After a median follow-up of 61.6 m, 44 HL progression events occurred: 21 NFT, 9 IFRT and 14 PET-positive. No patient received salvage treatment without documented progression. Only 5 HL-related deaths occurred (1 IFRT, 4 PET-positive), and 12 non-HL deaths (4 NFT, 6 IFRT, 2 PET-positive).1 For patients with CMR (N = 419), there was a strong association between MTD and EFS (hazard ratio [HR], 1.19; 95% confidence interval [CI], 1.02-1.39; P = .02), adjusting for treatment group, with an approximate 19% increase in HL risk per centimeter increase in MTD. The association was similar in both treatment groups (NFT HR, 1.20 [95% CI, 0.99-1.44; P = .06]; IFRT HR, 1.19 [95% CI, 0.92-1.55; P = .19]). The observed effect sizes did not markedly change after adjusting for baseline clinical risk factors, and similar results were observed for PFS (supplemental Table 1). In contrast, for PET-positive patients, there was no association between MTD and EFS (HR, 0.88; 95% CI, 0.70-1.11; P = .29) or PFS (HR, 0.87; 95% CI, 0.70-1.08; P = .21). In an exploratory analysis within the NFT group, MTD was dichotomized using increasing 1-cm intervals to investigate the relationship between MTD thresholds and EFS. The largest effect size was observed with an MTD threshold of ≥5 cm (Table 1). Similar results were observed for PFS; this threshold also performed best in time-dependent receiver operating characteristic curve analyses. It was not possible to assess MTD thresholds in the IFRT group with only 9 events. Among all randomized patients, 79 (18.9%) had MTD of ≥5 cm, the majority with mediastinal (n = 43), supraclavicular (n = 17), or cervical (n = 16) locations. Five-year EFS for patients with MTD of ≥5 cm randomly assigned to NFT and IFRT was 79.3% (n = 39; 95% CI, 66.6%-92.0%) and 94.9% (n = 40; 95% CI, 88.0%-100%), respectively (P = .03; Figure 1).
- Published
- 2020
- Full Text
- View/download PDF
40. Kinase inhibitors in CLL: drawing the roadmap
- Author
-
Nathan Fowler
- Subjects
Male ,Class I Phosphatidylinositol 3-Kinases ,Gastrointestinal Diseases ,Immunology ,MEDLINE ,Antineoplastic Agents ,Kaplan-Meier Estimate ,Heterocyclic Compounds, 4 or More Rings ,Biochemistry ,Phosphatidylinositol 3-Kinases ,Piperidines ,Agammaglobulinaemia Tyrosine Kinase ,Humans ,Medicine ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,Lymphoid Neoplasia ,Kinase ,business.industry ,Adenine ,Cell Biology ,Hematology ,Middle Aged ,Leukemia, Lymphocytic, Chronic, B-Cell ,Progression-Free Survival ,Neoplasm Proteins ,Cardiovascular Diseases ,Drug Resistance, Neoplasm ,Cancer research ,Female ,Drug Eruptions ,business - Abstract
Intolerance is the most common reason for kinase inhibitor (KI) discontinuation in chronic lymphocytic leukemia (CLL). Umbralisib, a novel highly selective phosphatidylinositol 3-kinase δ (PI3Kδ)/CK1ε inhibitor, is active and well tolerated in CLL patients. In this phase 2 trial (NCT02742090), umbralisib was initiated at 800 mg/d in CLL patients requiring therapy, who were intolerant to prior BTK inhibitor (BTKi) or PI3K inhibitor (PI3Ki) therapy, until progression or toxicity. Primary end point was progression‐free survival (PFS). Secondary end points included time to treatment failure and safety. DNA was genotyped for CYP3A4, CYP3A5, and CYP2D6 polymorphisms. Fifty-one patients were enrolled (44 BTKi intolerant and 7 PI3Kδi intolerant); median age was 70 years (range, 48‐96), with a median of 2 prior lines of therapy (range, 1‐7), 24% had del17p and/or TP53 mutation, and 65% had unmutated IGHV. Most common adverse events (AEs) leading to prior KI discontinuation were rash (27%), arthralgia (18%), and atrial fibrillation (16%). Median PFS was 23.5 months (95% CI, 13.1–not estimable), with 58% of patients on umbralisib for a longer duration than prior KI. Most common (≥5%) grade ≥3 AEs on umbralisib (all causality) were neutropenia (18%), leukocytosis (14%), thrombocytopenia (12%), pneumonia (12%), and diarrhea (8%). Six patients (12%) discontinued umbralisib because of an AE. Eight patients (16%) had dose reductions and were successfully rechallenged. These are the first prospective data to confirm that switching from a BTKi or alternate PI3Ki to umbralisib in this BTKi- and PI3Ki–intolerant CLL population can result in durable well-tolerated responses.
- Published
- 2021
- Full Text
- View/download PDF
41. Peri-CAR-T practice patterns and survival predictors for all CAR-T patients and post-CAR-T failure in aggressive B-NHL.
- Author
-
Zurko J, Nizamuddin I, Epperla N, David K, Cohen JB, Moyo TK, Ollila T, Hess B, Roy I, Ferdman R, Liu J, Chowdhury SM, Romancik J, Bhansali RS, Harris EI, Sorrell M, Masel R, Kittai AS, Denlinger N, Sigmund AM, Fitzgerald L, Galvez C, Ma S, Winter J, Pro B, Gordon LI, Danilov A, Stephens D, Shah NN, Kenkre V, Barta SK, Torka P, Shouse G, and Karmali R
- Subjects
- Humans, Retrospective Studies, Immunotherapy, Adoptive methods, Progression-Free Survival, Receptors, Chimeric Antigen therapeutic use, Lymphoma, B-Cell
- Abstract
Most patients receiving chimeric antigen receptor T-cell therapy (CAR-T) for aggressive B-cell non-Hodgkin lymphoma (B-NHL) do not experience a durable remission. Several novel agents are approved to treat relapsed, refractory aggressive B-NHL; however, it remains unclear how to sequence these therapies pre- and post-CAR-T. We conducted a multicenter retrospective analysis to describe peri-CAR-T practice patterns and survival predictors for patients receiving CD19-directed CAR-T. Patients (n = 514) from 13 centers treated with CAR-T for B-NHL between 2015-2021 were included in the study. Survival curves were constructed using Kaplan-Meier method. Multivariate Cox regression analysis was used to determine the impact of the variables on survival outcomes. For all patients receiving CAR-T, a greater number of lines of therapy pre-CAR-T apheresis and bridging therapy were predictive of inferior progression-free survival (PFS) and overall survival (OS). The median PFS and OS from the time of CAR-T cell infusion were 7.6 and 25.6 months, respectively. From the time of progression post-CAR-T, the median OS was 5.5 months. The median PFS of treatments administered in the first-line post-CAR-T failure was 2.8 months. Patients with refractory disease on day 30 had inferior OS and were less likely to receive subsequent treatment(s) than other patients with CAR-T failure. Allogeneic hematopoietic cell transplantation for selected patients at any time following CAR-T failure led to durable responses in over half of patients at 1 year. These data provide a benchmark for future clinical trials in patients with post-CAR-T cell progression, which remains an unmet clinical need., (© 2023 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
- 2023
- Full Text
- View/download PDF
42. van Rhee F, Rosenthal A, Kanhai K, et al. Siltuximab is associated with improved progression-free survival in idiopathic multicentric Castleman disease. Blood Adv. 2022;6(16):4773-4781.
- Subjects
- Humans, Progression-Free Survival, Antibodies, Monoclonal, Castleman Disease drug therapy
- Published
- 2023
- Full Text
- View/download PDF
43. Minimal residual disease is an independent predictor for 10-year survival in CLL.
- Author
-
Kwok, Marwan, Rawstron, Andy C., Varghese, Abraham, Evans, Paul A. S., O'Connor, Sheila J. M., Doughty, Chi, Newton, Darren J., Moreton, Paul, and Hillmen, Peter
- Subjects
- *
LYMPHOCYTIC leukemia , *IMMUNOTHERAPY , *PROGRESSION-free survival , *BONE marrow , *CYTOGENETICS - Abstract
Minimal residual disease (MRD) negativity, defined as <1 chronic lymphocytic leukemia (CLL) cell detectable per 10 000 leukocytes, has been shown to independently predict for clinical outcome in patients receiving combination chemoimmunotherapy in the frontline setting. However, the long-term prognostic value of MRD status in other therapeutic settings remains unclear. Here,weretrospectively analyzed, withupto 18 years follow-up, all patients at our institution who achieved at least a partial response (PR) with various therapies between 1996 and 2007, and received a bone marrow MRD assessment at the end of treatment according to the international harmonized approach. MRD negativity correlated with both progression-free survival (PFS) and overall survival (OS) independent of the type and line of treatment, as well as known prognostic factors including adverse cytogenetics. The greatest impact of achieving MRD negativity was seen in patients receiving frontline treatment, with 10-year PFS of 65% vs 10% and 10-year OS of 70% vs 30% for MRD-negative vs MRD-positive patients, respectively. Our results demonstrate the long-term benefit of achieving MRD negativity, regardless of the therapeutic setting and treatment modality, and support its use as a prognostic marker for long-term PFS and as a potential therapeutic goal in CLL. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
44. A trial of unrelated donor marrow transplantation for children with severe sickle cell disease.
- Author
-
Shalini Shenoy, Eapen, Mary, Panepinto, Julie A., Logan, Brent R., Juan Wu, Abraham, Allistair, Brochstein, Joel, Chaudhury, Sonali, Godder, Kamar, Haight, Ann E., Kasow, Kimberly A., Leung, Kathryn, Andreansky, Martin, Bhatia, Monica, Dalal, Jignesh, Haines, Hilary, Jaroscak, Jennifer, Lazarus, Hillard M., Levine, John E., and Krishnamurti, Lakshmanan
- Subjects
- *
SICKLE cell anemia in children , *BONE marrow transplantation , *BLOOD diseases , *GRAFT versus host disease , *PROGRESSION-free survival - Abstract
Children with sickle cell disease experience organ damage, impaired quality of life, and premature mortality. Allogeneic bone marrow transplant from an HLA-matched sibling can halt disease progression but is limited by donor availability. A Blood and Marrow Transplant Clinical Trials Network (BMT CTN) phase 2 trial conducted from 2008 to 2014 enrolled 30 children aged 4 to 19 years; 29 were eligible for evaluation. The primary objective was 1-year event-free survival (EFS) after HLA allele-matched (at HLA-A, -B, -C, and -DRB1 loci) unrelated donor transplant. The conditioning regimen included alemtuzumab, fludarabine, and melphalan. Graft-versus-host disease (GVHD) prophylaxis included calcineurin inhibitor, short-course methotrexate, and methylprednisolone. Transplant indications included stroke (n = 12), transcranial Doppler velocity >200 cm/s (n = 2), ≥3 vaso-occlusive pain crises per year (n = 12), or ≥2 acute chest syndrome episodes (n = 4) in the 2 years preceding enrollment. Median follow-up was 26 months (range, 12-62 months); graft rejection was 10%. The 1- and 2-year EFS rates were 76% and 69%, respectively. The corresponding rates for overall survival were 86% and 79%. The day 100 incidence rate of grade II-IV acute GVHD was 28%, and the 1-year incidence rate of chronic GVHD was 62%; 38% classified as extensive. There were 7 GVHD-related deaths. A 34% incidence of posterior reversible encephalopathy syndrome was noted in the first 6 months. Although the 1-year EFS met the prespecified target of ≥75%, this regimen cannot be considered sufficiently safe for widespread adoption without modifications to achieve more effective GVHD prophylaxis. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
45. Gene expression--based discovery of atovaquone as a STAT3 inhibitor and anticancer agent.
- Author
-
Xiang, Michael, Haesook Ki, Ho, Vincent T., Walker, Sarah R., Bar-Natan, Michal, Anahtar, Melodi, Suhu Liu, Toniolo, Patricia A., Kroll, Yasmin, Jones, Nichole, Giaccone, Zachary T., Heppler, Lisa N., Ye, Darwin Q., Marineau, Jason J., Shaw, Daniel, Bradner, James E., Blonquist, Traci, Neuberg, Donna, Hetz, Claudio, and Stone, Richard M.
- Subjects
- *
ATOVAQUONE , *ANTINEOPLASTIC agents , *TRANSCRIPTION factors , *GENETIC transcription , *GENE expression , *ACUTE myeloid leukemia treatment , *PROGRESSION-free survival - Abstract
The oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) is frequently activated inappropriately in a wide range of hematological and solid cancers, but clinically available therapies targeting STAT3 are lacking. Using a computational strategy to identify compounds opposing the gene expression signature of STAT3, we discovered atovaquone (Mepron), an antimicrobial approved by the US Food and Drug Administration, to be a potent STAT3 inhibitor. We show that, at drug concentrations routinely achieved clinically in human plasma, atovaquone inhibits STAT3 phosphorylation, the expression of STAT3 target genes, and the viability of STAT3-dependent hematological cancer cells. These effects were also observed with atovaquone treatment of primary blasts isolated from patients with acute myelogenous leukemia or acute lymphocytic leukemia. Atovaquone is not a kinase inhibitor but instead rapidly and specifically downregulates cell-surface expression of glycoprotein 130, which is required for STAT3 activation in multiple contexts. The administration of oral atovaquone to mice inhibited tumor growth and prolonged survival in a murine model of multiple myeloma. Finally, in patients with acute myelogenous leukemia treated with hematopoietic stem cell transplantation, extended use of atovaquone for Pneumocystis prophylaxis was associated with improved relapse-free survival. These findings establish atovaquone as a novel, clinically accessible STAT3 inhibitor with evidence of anticancer efficacy in both animal models and humans. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
46. Lenalidomide-bendamustine-rituximab in patients older than 65 years with untreated mantle cell lymphoma.
- Author
-
Albertsson-Lindblad, Alexandra, Kolstad, Arne, Laurell, Anna, Räty, Riikka, Grønbæk, Kirsten, Sundberg, Jan, Pedersen, Lone Bredo, Ralfkiær, Elisabeth, Karjalainen-Lindsberg, Marja-Liisa, Sundström, Christer, Ehinger, Mats, Geisler, Christian, and Jerkeman, Mats
- Subjects
- *
MANTLE cell lymphoma , *RITUXIMAB , *BIOTHERAPY , *PROGRESSION-free survival , *PNEUMOCYSTIS pneumonia , *CYTOMEGALOVIRUS retinitis , *THERAPEUTICS - Abstract
For elderly patients with mantle cell lymphoma (MCL), there is no defined standard therapy. In this multicenter, open-label phase 1/2 trial, we evaluated the addition of lenalidomide (LEN) to rituximab-bendamustine (R-B) as first-line treatment for elderly patients with MCL. Patients >65 years with untreated MCL, stages II-IV were eligible for inclusion. Primary end points were maximally tolerable dose (MTD) of LEN and progression-free survival (PFS). Patients received 6 cycles every four weeks of L-B-R (L D1-14, B 90 mg/m2 IV, days 1-2 and R 375 mg/m2 IV, day 1) followed by single LEN (days 1-21, every four weeks, cycles 7-13). Fifty-one patients (median age 71 years) were enrolled from 2009 to 2013. In phase 1, the MTD of LEN was defined as 10 mg in cycles 2 through 6, and omitted in cycle 1. After 6 cycles, the complete remission rate (CRR) was 64%, and 36% were MRD negative. At a median follow-up time of 31 months, median PFS was 42 months and 3-year overall survival was 73%. Infection was the most common nonhematologic grade 3 to 5 event and occurred in 21 (42%) patients. Opportunistic infections occurred in 3 patients: 2 Pneumocystis carinii pneumonia and 1 cytomegalovirus retinitis. Second primary malignancies (SPM) were observed in 8 patients (16%). LEN could safely be combined with R-B when added from the second cycle in patients with MCL, and was associated with a high rate of CR and molecular remission. However, we observed a high degree of severe infections and an unexpected high number of SPMs, which may limit its use. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
47. Brentuximab vedotin and AVD followed by involved-site radiotherapy in early stage, unfavorable risk Hodgkin lymphoma.
- Author
-
Kumar, Anita, Casulo, Carla, Yahalom, Joachim, Schöder, Heiko, Barr, Paul M., Caron, Philip, Chiu, April, Constine, Louis S., Drullinsky, Pamela, Friedberg, Jonathan W., Gerecitano, John F., Hamilton, Audrey, Hamlin, Paul A., Horwitz, Steven M., Jacob, Alexandra G., Matasar, Matthew J., McArthur, Gianna N., McCall, Susan J., Moskowitz, Alison J., and Noy, Ariela
- Subjects
- *
HODGKIN'S disease , *RADIOTHERAPY , *LYMPHOMAS , *POSITRON emission tomography , *PROGRESSION-free survival , *HYPERTENSION - Abstract
This multicenter pilot study assessed the safety and efficacy of brentuximab vedotin (BV) and AVD (adriamycin, vinblastine, and dacarbazine) followed by 30 Gy involved site radiation therapy (ISRT). Patients with newly diagnosed, early stage classical Hodgkin lymphoma (HL) with unfavorable-risk features were treated with 4 cycles of BV and AVD. Patients who achieved a negative positron emission tomography (PET) scan (Deauville score of 1-3) received 30 Gy ISRT. Thirty patients received treatment and were assessable for toxicity. Twenty-nine patients completed 4 cycles of BV 1 AVD, and 25 patients BV 1 AVD 1 30 Gy ISRT. No clinically significant noninfectious pneumonitis was observed. Serious adverseevents(≥grade 3)werereportedin4patients, includingfebrileneutropenia, peripheral neuropathy, and hypertension. After 2 and 4 cycles of BV 1 AVD, 90% (26 of 29) and 93% (27 or 29) of patients achieved a negative PET scan, respectively. Twopatientswith biopsy-proven primary refractory HL were treated off-study. All 25 patients who completed BV 1 AVD 1 ISRT achieved a complete response. With a median follow-up of 18.8 months, by intent to treat, the 1-year progression-free survival is 93.3% (95% confidence interval, 84-102). Overall, the treatment was well-tolerated with no evidence of significant pulmonary toxicity. The majority of patients (≥90%) achieved negative interim PET scans after 2 and 4 cycles of BV1AVD. Excludingthe 2 primary refractory patients, all patients are disease free, suggesting that this is a highly active treatment program even in patients with substantial disease bulk. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
48. Clinical efficacy of daratumumab monotherapy in patients with heavily pretreated relapsed or refractory multiple myeloma.
- Author
-
Usmani, Saad Z., Weiss, Brendan M., Plesner, Torben, Bahlis, Nizar J., Belch, Andrew, Lonial, Sagar, Lokhorst, Henk M., Voorhees, Peter M., Richardson, Paul G., Chari, Ajai, Sasser, A. Kate, Axel, Amy, Feng, Huaibao, Uhlar, Clarissa M., Jianping Wang, Khan, Imran, Ahmadi, Tahamtan, and Nahi, Hareth
- Subjects
- *
DRUG efficacy , *MULTIPLE myeloma treatment , *PROTEASOME inhibitors , *PROGRESSION-free survival , *IMMUNOSUPPRESSION , *ASPIRATION pneumonia - Abstract
The efficacy and favorable safety profile of daratumumab monotherapy in multiple myeloma (MM) was previously reported. An updated pooled analysis of 148 patients treated with daratumumab 16 mg/kg is presented. Data were combined from part 2 of a "first-in-human," phase 1/2 study of patients who relapsed after or were refractory to ⩾2 prior therapies and a phase 2 study of patients previously treated with ⩾3 prior lines of therapy (including a proteasome inhibitor [PI] and immunomodulatory drug [IMiD]) or were double refractory. Among the pooled population, patients received a median (range) of 5 (2-14) prior lines of therapy, and 86.5% were double refractory to a PI and IMID. Overall response rate was 31.1%, including 13 very good partial responses, 4 complete responses, and 3 stringent complete responses. The median duration of response was 7.6 months (95% confidence interval [CI], 5.6-not evaluable [NE]). The median progression-free survival (PFS) and overall survival (OS) was 4.0 months (95% CI, 2.8-5.6) and 20.1 months (95% CI, 16.6-NE), respectively. When stratified by responders versus stable disease/minimal response versus progressive disease/NE, median PFS was 15.0 months (95% CI, 7.4-NE) versus 3.0 months (95% CI, 2.8-3.7) versus 0.9 months (95% CI, 0.9-1.0), respectively, and median OS was NE (95% CI, NE-NE) versus 18.5 months (95% CI, 15.1-22.4) versus 3.7 months (95% CI, 1.7-7.6), respectively. No new safety signals were identified. In this pooled dataset, daratumumab 16 mg/kg monotherapy demonstrated rapid, deep, and durable responses, with a clinical benefit that extended to patients with stable disease or better. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
49. CHAMPION-1: a phase 1/2 study of once-weekly carfilzomib and dexamethasone for relapsed or refractory multiple myeloma.
- Author
-
Berenson, James R., Cartmell, Alan, Bessudo, Alberto, Lyons, Roger M., Harb, Wael, Tzachanis, Dimitrios, Agajanian, Richy, Boccia, Ralph, Coleman, Morton, Moss, Robert A., Rifkin, Robert M., Patel, Priti, Dixon, Sandra, Ying Ou, Anderl, Janet, Aggarwal, Sanjay, and Berdeja, Jesus G.
- Subjects
- *
PROTEASOME inhibitors , *DEXAMETHASONE , *MYELOMA proteins , *PROGRESSION-free survival , *DISEASE relapse - Abstract
Carfilzomib, a proteasome inhibitor, is approved in the United States as a single agent, and in combination with dexamethasone or lenalidomide/dexamethasone (KRd) for relapsed or refractory multiple myeloma (MM). Under the single-agent and KRd approvals, carfilzomib is administered as a 10-minute IV infusion on days 1, 2, 8, 9, 15, and 16 of 28- day cycles (20mg/m2 [cycle 1, days 1-2]; 27mg/m2 thereafter).Thismulticenter, single-arm, phase 1/2 study, Community Harmonized Assessment of Myeloma Patients via an Integrated Oncology Network-1 (CHAMPION-1), evaluated once-weekly carfilzomib with dexamethasone in relapsed, or relapsed and refractory MM (1-3 prior therapies). Patients received carfilzomib (30-minute IVinfusion) on days 1, 8, and15 of 28-day cycles. Thephase 1 portion used a 3 1 3 dose-escalation scheme to determine the maximum tolerated dose (MTD) of carfilzomib. During phase 2, patients received carfilzomib on the same schedule at theMTD. Patients received dexamethasone (40mg) on days 1, 8, 15, and 22; dexamethasone was omitted on day 22 for cycles 91. A total of 116 patients were enrolled. The MTD was 70 mg/m2, and 104 patients (phase 1/2) received carfilzomib 70 mg/m2. At 70 mg/m2, the median number of prior regimens was 1; and 52% were bortezomib-refractory. At 70 mg/m2, the most common grade ≥3 adverse events were fatigue (11%) and hypertension (7%). Overall response rate at 70 mg/m2 was 77%. Median progression-free survival was 12.6 months. These findings merit additional evaluation of the once-weekly dosing regimen. This trial was registered at www.clinicaltrials. gov as #NCT01677858. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
50. Dexamethasone vs prednisone in induction treatment of pediatric ALL: results of the randomized trial AIEOP-BFM ALL 2000.
- Author
-
Möricke, Anja, Zimmermann, Martin, Valsecchi, Maria Grazia, Stanulla, Martin, Biondi, Andrea, Mann, Georg, Locatelli, Franco, Cazzaniga, Giovanni, Niggli, Felix, Aricò, Maurizio, Bartram, Claus R., Attarbaschi, Andishe, Silvestri, Daniela, Beier, Rita, Basso, Giuseppe, Ratei, Richard, Kulozik, Andreas E., Lo Nigro, Luca, Kremens, Bernhard, and Greiner, Jeanette
- Subjects
- *
PREDNISONE , *DEXAMETHASONE , *LYMPHOBLASTIC leukemia , *CANCER relapse , *PROGRESSION-free survival - Abstract
Induction therapy for childhood acute lymphoblastic leukemia (ALL) traditionally includes prednisone; yet, dexamethasone may have higher antileukemic potency, leading to fewer relapses and improved survival. After a 7-day prednisone prephase, 3720 patients enrolled on trial Associazione Italiana di Ematologia e Oncologia Pediatrica and Berlin-Frankfurt-Münster (AIEOP-BFM) ALL 2000 were randomly selected to receive either dexamethasone (10 mg/m2 per day) or prednisone (60 mg/m2 per day) for 3 weeks plus tapering in induction. The 5-year cumulative incidence of relapse (± standard error) was 10.8 ± 0.7% in the dexamethasone and 15.6 ± 0.8% in the prednisone group (P < .0001), showing the largest effect on extramedullary relapses. The benefit of dexamethasone was partially counterbalanced by a significantly higher induction-related death rate (2.5% vs 0.9%, P = .00013), resulting in 5-year event-free survival rates of 83.9 ± 0.9% for dexamethasone and 80.8 ± 0.9% for prednisone (P = .024). No difference was seen in 5-year overall survival (OS) in the total cohort (dexamethasone, 90.3 ± 0.7%; prednisone, 90.5 ± 0.7%). Retrospective analyses of predefined subgroups revealed a significant survival benefit from dexamethasone only for patients with T-cell ALL and good response to the prednisone prephase (prednisone good-response [PGR]) (dexamethasone, 91.4 ± 2.4%; prednisone, 82.6 ± 3.2%; P = .036). In patients with precursor B-cell ALL and PGR, survival after relapse was found to be significantly worse if patients were previously assigned to the dexamethasone arm. We conclude that, for patients with PGR in the large subgroup of precursor B-cell ALL, dexamethasone especially reduced the incidence of better salvageable relapses, resulting in inferior survival after relapse. This explains the lack of benefit from dexamethasone in overall survival that we observed in the total cohort except in the subset of T-cell ALL patients with PGR. This trial was registered at www.clinicaltrials.gov (BFM: NCT00430118, AIEOP: NCT00613457). [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.