Juan F. Combariza-Vallejo, Rocío Orduz-Rodriguez, Natalia Gomez-Lopera, Claudia C. Colmenares-Mejia, Fabián A. Mejía-Casadiego, Marcela Godoy-Corredor, Mario Arturo Isaza-Ruget
Mantle cell lymphoma is a type of B-cell lymphoma with limited epidemiologic data in Latin America. The main objective was evaluating survival, in mantle cell lymphoma in Colombia and the influence of hematopoietic stem cell transplantation in the outcomes. In a retrospective cohort of new diagnostic Mantle Cell Lymphoma between 2010 and 2024, we identified main characteristics at diagnosis and the influence of MIPI score and ASCT. We followed patients until progression or death and compared progression-free survival and overall survival. We included 139 patients; the median age was 64 years. For patients classified as low risk according to the MIPI score, the 36-month OS rate was 79.7% (95% CI: 68%–93.5%). Intermediate risk 51.5% (95% CI: 38.2%–69.4%). And High-risk 40.4% (95% CI: 26.9%–59.5%) (p < 0.001). In the group that underwent ASCT, the 36-month OS rate was 75.2% (95% CI: 60.1%–94.6%), and in the non-transplanted group was 49% (95% CI: 39.5%–60.7%), HR 0.38 (95% CI; 019–0.74) (p = 0.002). High-risk patients and those who were not transplanted had the lowest survival compared to the other categories. These findings underscore the need for risk stratification, and effective treatment strategies, including the HSCT patient population in Colombia.
{"title":"Survival Analysis in a Colombian Cohort of Mantle Cell Lymphoma. A Real-World Experience","authors":"Juan F. Combariza-Vallejo, Rocío Orduz-Rodriguez, Natalia Gomez-Lopera, Claudia C. Colmenares-Mejia, Fabián A. Mejía-Casadiego, Marcela Godoy-Corredor, Mario Arturo Isaza-Ruget","doi":"10.1002/hon.70085","DOIUrl":"https://doi.org/10.1002/hon.70085","url":null,"abstract":"<div>\u0000 \u0000 <p>Mantle cell lymphoma is a type of B-cell lymphoma with limited epidemiologic data in Latin America. The main objective was evaluating survival, in mantle cell lymphoma in Colombia and the influence of hematopoietic stem cell transplantation in the outcomes. In a retrospective cohort of new diagnostic Mantle Cell Lymphoma between 2010 and 2024, we identified main characteristics at diagnosis and the influence of MIPI score and ASCT. We followed patients until progression or death and compared progression-free survival and overall survival. We included 139 patients; the median age was 64 years. For patients classified as low risk according to the MIPI score, the 36-month OS rate was 79.7% (95% CI: 68%–93.5%). Intermediate risk 51.5% (95% CI: 38.2%–69.4%). And High-risk 40.4% (95% CI: 26.9%–59.5%) (<i>p</i> < 0.001). In the group that underwent ASCT, the 36-month OS rate was 75.2% (95% CI: 60.1%–94.6%), and in the non-transplanted group was 49% (95% CI: 39.5%–60.7%), HR 0.38 (95% CI; 019–0.74) (<i>p</i> = 0.002). High-risk patients and those who were not transplanted had the lowest survival compared to the other categories. These findings underscore the need for risk stratification, and effective treatment strategies, including the HSCT patient population in Colombia.</p>\u0000 </div>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143857133","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pallawi Torka, Natalie S. Grover, Timothy J. Voorhees, Reem Karmali, Kaitlin Annunzio, Marcus P. Watkins, Andrea Anampa-Guzmán, Heather Reves, Montreh Tavakkoli, Beth Christian, Colin Thomas, Stefan K. Barta, Praveen Ramakrishnan Geethakumari, Nancy L. Bartlett, Geoffrey Shouse, Adam J. Olszewski, Narendranath Epperla
Given the paucity of age-specific data about biology, presentation, and treatment outcomes in adults with MZL, we sought to evaluate differences between younger (≤ 70 years) and older (> 70 years) patients with MZL in a large retrospective cohort treated in the contemporary era (2010 onwards). The primary objective was progression-free survival (PFS), while secondary objectives included the evaluation of overall survival (OS) and the cumulative incidence of transformation between the 2 groups. A total of 598 patients were included in the analysis and among these 32% were > 70 years of age. There were no age-based differences in the prevalence of NMZL, SMZL, and EMZL. Older patients had a higher incidence of adverse prognostic features at diagnosis such as worse performance status, advanced stage disease, and bone marrow involvement, yet were more likely to be treated with single-agent rituximab than chemoimmunotherapy. Age > 70 years was associated with inferior PFS and OS after controlling for clinically relevant risk factors and accounting for differences in first-line treatment. Receipt of rituximab monotherapy was associated with significantly inferior PFS overall, however, the type of first-line therapy did not impact OS in any group. Our data suggests that despite the development of new drugs for MZL, age remains an independent predictor of inferior outcomes. Investigation of targeted therapy combinations in the first-line setting may yield the required balance of efficacy and toxicity in older adults with MZL.
{"title":"Impact of Age on Biology, Presentation and Outcomes in Marginal Zone Lymphoma: Results From a Multicenter Cohort Study","authors":"Pallawi Torka, Natalie S. Grover, Timothy J. Voorhees, Reem Karmali, Kaitlin Annunzio, Marcus P. Watkins, Andrea Anampa-Guzmán, Heather Reves, Montreh Tavakkoli, Beth Christian, Colin Thomas, Stefan K. Barta, Praveen Ramakrishnan Geethakumari, Nancy L. Bartlett, Geoffrey Shouse, Adam J. Olszewski, Narendranath Epperla","doi":"10.1002/hon.70087","DOIUrl":"https://doi.org/10.1002/hon.70087","url":null,"abstract":"<p>Given the paucity of age-specific data about biology, presentation, and treatment outcomes in adults with MZL, we sought to evaluate differences between younger (≤ 70 years) and older (> 70 years) patients with MZL in a large retrospective cohort treated in the contemporary era (2010 onwards). The primary objective was progression-free survival (PFS), while secondary objectives included the evaluation of overall survival (OS) and the cumulative incidence of transformation between the 2 groups. A total of 598 patients were included in the analysis and among these 32% were > 70 years of age. There were no age-based differences in the prevalence of NMZL, SMZL, and EMZL. Older patients had a higher incidence of adverse prognostic features at diagnosis such as worse performance status, advanced stage disease, and bone marrow involvement, yet were more likely to be treated with single-agent rituximab than chemoimmunotherapy. Age > 70 years was associated with inferior PFS and OS after controlling for clinically relevant risk factors and accounting for differences in first-line treatment. Receipt of rituximab monotherapy was associated with significantly inferior PFS overall, however, the type of first-line therapy did not impact OS in any group. Our data suggests that despite the development of new drugs for MZL, age remains an independent predictor of inferior outcomes. Investigation of targeted therapy combinations in the first-line setting may yield the required balance of efficacy and toxicity in older adults with MZL.</p>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70087","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143857145","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Relapse and refractory T-cell acute lymphoblastic leukemia and lymphoblastic lymphoma (R/R T-ALL/LBL) patients have poor outcomes. Chimeric antigen receptor (CAR)-T-cell therapy for T-ALL/LBL has shown encouraging results in the early stages of clinical trials. Here, we retrospectively analyzed the outcomes of 12 patients with R/R T-ALL/LBL who received CD7 CAR-T cell therapy. Eleven patients received autologous CAR-T cells, while one patient received allogeneic CAR-T cells. On Day 28 post-infusion, 67% (8/12) of patients achieved an overall response (ORR). At a median follow-up of 134 (14–925) days, the median overall survival (OS) was 134 days, and the progression-free survival (PFS) was 81 days. Among the 8 patients who achieved remission after CD7 CAR-T cell infusion, 5 patients received consolidation allogeneic hematopoietic stem cell transplantation (allo-HSCT). Compared with 3 patients who did not undergo consolidation allo-HSCT, patients with allo-HSCT as consolidation showed a trend toward better OS (allo-HSCT vs. control: 6-month OS, 60% vs. 33.3%, p = 0.073) and better PFS (allo-HSCT vs. control: 6-month PFS, 60% vs. 0%, p = 0.022). Cytokine release syndrome (CRS) occurred in all patients (grade 1–2 in 67% of patients, grade 3 in 33% of patients), and one patient experienced neurotoxicity. CD7 CAR-T cell therapy is a promising option for R/R T-ALL/LBL patients with manageable adverse events. Moreover, CD7 CAR-T cell infusion followed by consolidation allo-HSCT in R/R T-ALL/LBL patients might play an important role in prolonging remission duration.
{"title":"Clinical Outcomes of CD7 CAR-T Cell Therapy in Relapsed or Refractory T-Cell Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma Patients","authors":"Jinyan Xiao, Yujie Liu, Yifan Shen, Jiaqi Li, Ying Wang, Suning Chen, Xiaowen Tang, Lin Yang, Biqi Zhou, Yang Xu, Depei Wu","doi":"10.1002/hon.70081","DOIUrl":"https://doi.org/10.1002/hon.70081","url":null,"abstract":"<div>\u0000 \u0000 <p>Relapse and refractory T-cell acute lymphoblastic leukemia and lymphoblastic lymphoma (R/R T-ALL/LBL) patients have poor outcomes. Chimeric antigen receptor (CAR)-T-cell therapy for T-ALL/LBL has shown encouraging results in the early stages of clinical trials. Here, we retrospectively analyzed the outcomes of 12 patients with R/R T-ALL/LBL who received CD7 CAR-T cell therapy. Eleven patients received autologous CAR-T cells, while one patient received allogeneic CAR-T cells. On Day 28 post-infusion, 67% (8/12) of patients achieved an overall response (ORR). At a median follow-up of 134 (14–925) days, the median overall survival (OS) was 134 days, and the progression-free survival (PFS) was 81 days. Among the 8 patients who achieved remission after CD7 CAR-T cell infusion, 5 patients received consolidation allogeneic hematopoietic stem cell transplantation (allo-HSCT). Compared with 3 patients who did not undergo consolidation allo-HSCT, patients with allo-HSCT as consolidation showed a trend toward better OS (allo-HSCT vs. control: 6-month OS, 60% vs. 33.3%, <i>p</i> = 0.073) and better PFS (allo-HSCT vs. control: 6-month PFS, 60% vs. 0%, <i>p</i> = 0.022). Cytokine release syndrome (CRS) occurred in all patients (grade 1–2 in 67% of patients, grade 3 in 33% of patients), and one patient experienced neurotoxicity. CD7 CAR-T cell therapy is a promising option for R/R T-ALL/LBL patients with manageable adverse events. Moreover, CD7 CAR-T cell infusion followed by consolidation allo-HSCT in R/R T-ALL/LBL patients might play an important role in prolonging remission duration.</p>\u0000 </div>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143826877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Diffuse large B cell lymphoma (DLBCL) is a type of common and fatal non-Hodgkin lymphoma. This study aimed to detect the specific function of lncRNA ARAP1-AS1 in DLBCL. The in vitro experiments were performed using RT-qPCR, western blotting, MTT, colony formation, flow cytometry analysis, FISH, RNA pulldown, and luciferase reporter assays. A xenograft mouse model was used to evaluate tumor growth in vivo. ARAP1-AS1 expression was upregulated in DLBCL tissues and cells. ARAP1-AS1 knockdown inhibited DLBCL cell proliferation and promoted apoptosis. ARAP1-AS1 activated PI3K/AKT signaling by upregulating EMP1 expression via miR-508–5p. EMP1 overexpression markedly abolished the effect of ARAP1-AS1 knockdown on DLBCL cell proliferation and apoptosis, and PI3K inhibitor reversed the effect of ARAP1-AS1 overexpression on DLBCL cells. ARAP1-AS1 knockdown inhibited DLBCL tumor growth and reduced Ki-67, EMP1, and p-AKT expression in xenograft mouse models. ARAP1-AS1 knockdown exerts anti-tumor effect on DLBCL progression through the miR-508–5p/EMP1/PI3K/AKT axis.
{"title":"ARAP1-AS1 Overexpression Increases Diffuse Large B Cell Lymphoma Progression by Sponging miR-508–5p to Activate the EMP1-PI3K/AKT Pathway","authors":"Yamei Chen, Minmin Ren, Lei Zhu, Ting Sun, Xiaoyong Wang, Lingling Jiang, Linlin Zhou, Dongyun Gao","doi":"10.1002/hon.70056","DOIUrl":"https://doi.org/10.1002/hon.70056","url":null,"abstract":"<div>\u0000 \u0000 <p>Diffuse large B cell lymphoma (DLBCL) is a type of common and fatal non-Hodgkin lymphoma. This study aimed to detect the specific function of lncRNA ARAP1-AS1 in DLBCL. The in vitro experiments were performed using RT-qPCR, western blotting, MTT, colony formation, flow cytometry analysis, FISH, RNA pulldown, and luciferase reporter assays. A xenograft mouse model was used to evaluate tumor growth in vivo. ARAP1-AS1 expression was upregulated in DLBCL tissues and cells. ARAP1-AS1 knockdown inhibited DLBCL cell proliferation and promoted apoptosis. ARAP1-AS1 activated PI3K/AKT signaling by upregulating EMP1 expression via miR-508–5p. EMP1 overexpression markedly abolished the effect of ARAP1-AS1 knockdown on DLBCL cell proliferation and apoptosis, and PI3K inhibitor reversed the effect of ARAP1-AS1 overexpression on DLBCL cells. ARAP1-AS1 knockdown inhibited DLBCL tumor growth and reduced Ki-67, EMP1, and <i>p</i>-AKT expression in xenograft mouse models. ARAP1-AS1 knockdown exerts anti-tumor effect on DLBCL progression through the miR-508–5p/EMP1/PI3K/AKT axis.</p>\u0000 </div>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143826876","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Robin Noel, Catalina Montes de Oca, Patrick Sfumato, Anne Calleja, Aude Collignon, Ferdinand Villetard, Jean Marc Schiano de Collela, Frédéric Benizri, Pilar Dutari, Cécile Castoldi, Faezeh Izadifar-Legrand, Luca Inchiappa, Catherine Evans, Gabriel Brisou, Lenaig Mescam, Vincent Camus, Thibaut Reichert
Immunochemotherapy has demonstrated high cure rates in primary mediastinal large B-cell lymphoma (PMBCL). Recent findings from the IELSG 37 study show that certain specific dose-dense treatment regimens are more likely to eliminate the need for consolidative radiation therapy, thereby reducing long-term toxicity. Here, we report our experience with DA-R-EPOCH in a cohort of 70 patients treated at our cancer center. This retrospective, observational, single-center study includes patients treated between 2013 and 2021. Treatment response was evaluated using positron emission tomography-computed tomography (PET-CT) scans and assessed according to the Deauville Score. With a median follow-up of 36.4 months, 29% of patients achieved a complete metabolic response (CMR) on interim PET-CT, while 71% achieved CMR at the end of treatment. Despite these results, 3-year overall survival (OS) and progression-free survival (PFS) rates were 99% and 93%, respectively. Our findings highlight the importance of tailoring treatment regimens, allowing for reduced reliance on consolidative therapies in line with recent advancements. Notably, none of the patients who achieved CMR on interim PET-CT experienced relapse, and no disease progression was observed beyond 1 year after completing DA-R-EPOCH. Additionally, our results indicate that dose intensification beyond level 2 does not appear to improve survival, emphasizing the important implications for minimizing treatment-related toxicity. We also analyzed the management and outcomes of refractory and relapsed cases, underscoring the challenges in treating non-responding patients identified by interim PET-CT, the limited efficacy of salvage immunochemotherapy, and the interesting role of radiation therapy in these cases.
{"title":"Long-Term Outcomes and Management Strategies With DA-R-EPOCH in Primary Mediastinal B-Cell Lymphoma: Insights From a Single-Center Experience","authors":"Robin Noel, Catalina Montes de Oca, Patrick Sfumato, Anne Calleja, Aude Collignon, Ferdinand Villetard, Jean Marc Schiano de Collela, Frédéric Benizri, Pilar Dutari, Cécile Castoldi, Faezeh Izadifar-Legrand, Luca Inchiappa, Catherine Evans, Gabriel Brisou, Lenaig Mescam, Vincent Camus, Thibaut Reichert","doi":"10.1002/hon.70060","DOIUrl":"https://doi.org/10.1002/hon.70060","url":null,"abstract":"<p>Immunochemotherapy has demonstrated high cure rates in primary mediastinal large B-cell lymphoma (PMBCL). Recent findings from the IELSG 37 study show that certain specific dose-dense treatment regimens are more likely to eliminate the need for consolidative radiation therapy, thereby reducing long-term toxicity. Here, we report our experience with DA-R-EPOCH in a cohort of 70 patients treated at our cancer center. This retrospective, observational, single-center study includes patients treated between 2013 and 2021. Treatment response was evaluated using positron emission tomography-computed tomography (PET-CT) scans and assessed according to the Deauville Score. With a median follow-up of 36.4 months, 29% of patients achieved a complete metabolic response (CMR) on interim PET-CT, while 71% achieved CMR at the end of treatment. Despite these results, 3-year overall survival (OS) and progression-free survival (PFS) rates were 99% and 93%, respectively. Our findings highlight the importance of tailoring treatment regimens, allowing for reduced reliance on consolidative therapies in line with recent advancements. Notably, none of the patients who achieved CMR on interim PET-CT experienced relapse, and no disease progression was observed beyond 1 year after completing DA-R-EPOCH. Additionally, our results indicate that dose intensification beyond level 2 does not appear to improve survival, emphasizing the important implications for minimizing treatment-related toxicity. We also analyzed the management and outcomes of refractory and relapsed cases, underscoring the challenges in treating non-responding patients identified by interim PET-CT, the limited efficacy of salvage immunochemotherapy, and the interesting role of radiation therapy in these cases.</p>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70060","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143824729","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Danai Dima, Utkarsh Goel, Fauzia Ullah, Beth Faiman, Diana Basali, Sandra Mazzoni, Louis S. Williams, Christy Samaras, Jason Valent, Faiz Anwer, Jack Khouri, Shahzad Raza
Localized light chain amyloidosis (loc-AL) is a rare disorder characterized by localized deposition of misfolded AL fibrils. There are limited data on patterns of disease presentation and long-term outcomes. In this study, we retrospectively reviewed 146 patients with loc-AL at our institution between January 1, 2010, and March 1, 2024. We excluded patients with evidence of systemic AL amyloidosis. We calculated local (PFSL) and systemic (PFSs) progression free survival (PFS) and overall survival (OS) using the Kaplan-Meier method. We found that loc-AL most commonly involved the respiratory (26%), gastrointestinal (17%), head and neck (17%) and genitourinary (10%) systems. Overall, 51% patients were asymptomatic at presentation, and 16% had a co-existent autoimmune disease. First line management included observation (52%), surgical resection (39%), chemotherapy (3%), and radiation (2%). Most patients (59%) had a response with first-line therapy. The median PFSL was ∼15 years (10-year PFSL 68%), and median OS was not reached (10-year OS 83%). None of the patients had progression to systemic amyloidosis. Seventeen patients had local recurrence and required second line therapy. In conclusion, loc-AL has an excellent prognosis and does not progress to systemic AL amyloidosis. Observation and/or surgical removal are usually adequate first-line approaches; however, a small proportion of patients can relapse locally requiring repeated interventions for symptom control.
{"title":"Presentation and Outcomes of Localized Immunoglobulin Light Chain Amyloidosis: 14-Year Experience of an Academic Center","authors":"Danai Dima, Utkarsh Goel, Fauzia Ullah, Beth Faiman, Diana Basali, Sandra Mazzoni, Louis S. Williams, Christy Samaras, Jason Valent, Faiz Anwer, Jack Khouri, Shahzad Raza","doi":"10.1002/hon.70082","DOIUrl":"https://doi.org/10.1002/hon.70082","url":null,"abstract":"<p>Localized light chain amyloidosis (loc-AL) is a rare disorder characterized by localized deposition of misfolded AL fibrils. There are limited data on patterns of disease presentation and long-term outcomes. In this study, we retrospectively reviewed 146 patients with loc-AL at our institution between January 1, 2010, and March 1, 2024. We excluded patients with evidence of systemic AL amyloidosis. We calculated local (PFS<sub>L</sub>) and systemic (PFS<sub>s</sub>) progression free survival (PFS) and overall survival (OS) using the Kaplan-Meier method. We found that loc-AL most commonly involved the respiratory (26%), gastrointestinal (17%), head and neck (17%) and genitourinary (10%) systems. Overall, 51% patients were asymptomatic at presentation, and 16% had a co-existent autoimmune disease. First line management included observation (52%), surgical resection (39%), chemotherapy (3%), and radiation (2%). Most patients (59%) had a response with first-line therapy. The median PFS<sub>L</sub> was ∼15 years (10-year PFS<sub>L</sub> 68%), and median OS was not reached (10-year OS 83%). None of the patients had progression to systemic amyloidosis. Seventeen patients had local recurrence and required second line therapy. In conclusion, loc-AL has an excellent prognosis and does not progress to systemic AL amyloidosis. Observation and/or surgical removal are usually adequate first-line approaches; however, a small proportion of patients can relapse locally requiring repeated interventions for symptom control.</p>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70082","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143824730","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lucio N. Gordan, Rohan Medhekar, Alex Z. Fu, Mostafa Shokoohi, Abril Oliva Ramirez, Nicolle Bonar, Bao-Ngoc Nguyen, Michaela Spence, Rebecca McTavish, Tim Disher, Santosh Gautam, Niodita Gupta-Werner, Shuchita Kaila, Anjan J. Patel
Daratumumab in combination with lenalidomide and dexamethasone (DRd) and bortezomib in combination with lenalidomide and dexamethasone (VRd) are guideline-recommended preferred regimens for initial treatment of transplant-ineligible (TIE) patients with newly diagnosed multiple myeloma (NDMM). This study aimed to systematically identify evidence on the clinical effectiveness of DRd and VRd as first-line treatments for patients with TIE NDMM and to conduct a meta-analysis. Ovid MEDLINE, Embase, and Cochrane Library were searched from January 2019 to June 2023, along with key congresses from January 2018 to June 2023. Bibliographies of relevant systematic literature reviews (SLR) were hand-searched. Randomized controlled trials and appropriately adjusted non-randomized studies comparing DRd versus VRd as first-line treatment for TIE NDMM were included. Overall, five records from three unique studies were identified. The fixed-effects meta-analysis showed a lower risk of disease progression or death with DRd versus VRd using the naïve approach (hazard ratio [HR]: 0.60; 95% confidence interval [CI]: 0.46, 0.77) as well as with the adjusted approach, which accounted for both double counting (i.e., two studies shared one comparison) and variance inflation due to studies with moderate and high risk of bias (HR: 0.56; 95% CI: 0.39, 0.82). In the absence of clinical trials with head-to-head comparison of these treatment regimens, these results could help inform the selection of optimal first-line treatment for TIE NDMM patients.
{"title":"Daratumumab, Lenalidomide, and Dexamethasone Versus Bortezomib, Lenalidomide, and Dexamethasone in Transplant-Ineligible Newly Diagnosed Multiple Myeloma: A Systematic Literature Review and Meta-Analysis","authors":"Lucio N. Gordan, Rohan Medhekar, Alex Z. Fu, Mostafa Shokoohi, Abril Oliva Ramirez, Nicolle Bonar, Bao-Ngoc Nguyen, Michaela Spence, Rebecca McTavish, Tim Disher, Santosh Gautam, Niodita Gupta-Werner, Shuchita Kaila, Anjan J. Patel","doi":"10.1002/hon.70061","DOIUrl":"https://doi.org/10.1002/hon.70061","url":null,"abstract":"<p>Daratumumab in combination with lenalidomide and dexamethasone (DRd) and bortezomib in combination with lenalidomide and dexamethasone (VRd) are guideline-recommended preferred regimens for initial treatment of transplant-ineligible (TIE) patients with newly diagnosed multiple myeloma (NDMM). This study aimed to systematically identify evidence on the clinical effectiveness of DRd and VRd as first-line treatments for patients with TIE NDMM and to conduct a meta-analysis. Ovid MEDLINE, Embase, and Cochrane Library were searched from January 2019 to June 2023, along with key congresses from January 2018 to June 2023. Bibliographies of relevant systematic literature reviews (SLR) were hand-searched. Randomized controlled trials and appropriately adjusted non-randomized studies comparing DRd versus VRd as first-line treatment for TIE NDMM were included. Overall, five records from three unique studies were identified. The fixed-effects meta-analysis showed a lower risk of disease progression or death with DRd versus VRd using the naïve approach (hazard ratio [HR]: 0.60; 95% confidence interval [CI]: 0.46, 0.77) as well as with the adjusted approach, which accounted for both double counting (i.e., two studies shared one comparison) and variance inflation due to studies with moderate and high risk of bias (HR: 0.56; 95% CI: 0.39, 0.82). In the absence of clinical trials with head-to-head comparison of these treatment regimens, these results could help inform the selection of optimal first-line treatment for TIE NDMM patients.</p>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70061","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143809780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chan Tze Wei, Hein Than, Feng-Ju Huang, Gauri Billa, Lai Heng Lee
Myelofibrosis (MF) is characterized by anemia, constitutional symptoms, hepatosplenomegaly and bone marrow fibrosis, and is associated with poor survival. The janus kinase inhibitor (JAKi) ruxolitinib has been the mainstay of treatment for over a decade. Despite demonstrated symptomatic and quality of life improvement, unmet clinical needs persist. A literature review identified promising novel targeted treatment options in MF using pre-set selection criteria (available Phase 2 or 3 data, minimum enrollment of 50 patients, trial end date within the last 5 years). Available data for novel and approved therapies were extracted, tabulated, and analyzed for clinical relevancy. From an initial shortlist of 48, 16 retained molecules were selected for inclusion. Other JAKi (pacritinib, momelotinib, jaktinib) address treatment-related cytopenia, expanding the therapeutic utility of this class of agents to patients with baseline anemia or thrombocytopenia. Novel candidates exploit multiple molecular pathways, and offer the potential to improve the management of MF-associated cytopenia (imetelstat, pelabresib, navitoclax, selinexor, luspatercept, sotatercept, elritercept, LCL161, bomedemstat) and recover bone marrow fibrosis (imetelstat, pelabresib, navitoclax and bomedemstat). It remains to be seen if these newer agents can induce any remission in MF and enable patients to come off therapy, but the future is beginning to look much brighter.
{"title":"Managing Myelofibrosis: Matching Advances in Treatments With Clinical Unmet Needs","authors":"Chan Tze Wei, Hein Than, Feng-Ju Huang, Gauri Billa, Lai Heng Lee","doi":"10.1002/hon.70053","DOIUrl":"https://doi.org/10.1002/hon.70053","url":null,"abstract":"<p>Myelofibrosis (MF) is characterized by anemia, constitutional symptoms, hepatosplenomegaly and bone marrow fibrosis, and is associated with poor survival. The janus kinase inhibitor (JAKi) ruxolitinib has been the mainstay of treatment for over a decade. Despite demonstrated symptomatic and quality of life improvement, unmet clinical needs persist. A literature review identified promising novel targeted treatment options in MF using pre-set selection criteria (available Phase 2 or 3 data, minimum enrollment of 50 patients, trial end date within the last 5 years). Available data for novel and approved therapies were extracted, tabulated, and analyzed for clinical relevancy. From an initial shortlist of 48, 16 retained molecules were selected for inclusion. Other JAKi (pacritinib, momelotinib, jaktinib) address treatment-related cytopenia, expanding the therapeutic utility of this class of agents to patients with baseline anemia or thrombocytopenia. Novel candidates exploit multiple molecular pathways, and offer the potential to improve the management of MF-associated cytopenia (imetelstat, pelabresib, navitoclax, selinexor, luspatercept, sotatercept, elritercept, LCL161, bomedemstat) and recover bone marrow fibrosis (imetelstat, pelabresib, navitoclax and bomedemstat). It remains to be seen if these newer agents can induce any remission in MF and enable patients to come off therapy, but the future is beginning to look much brighter.</p>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S1","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70053","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143787165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}