bims-hemali Biomed News
on Hematologic malignancies
Issue of 2025–04–27
eighteen papers selected by
Alexandros Alexandropoulos, Γενικό Νοσοκομείο Αθηνών Λαϊκό



  1. Biomed J. 2025 Apr 18. pii: S2319-4170(25)00028-9. [Epub ahead of print] 100854
      
    Keywords:  chronic myeloid leukemia; deep molecular response; hematopoietic stem cells; treatment-free remission; tyrosine kinase inhibitor
    DOI:  https://doi.org/10.1016/j.bj.2025.100854
  2. Am Soc Clin Oncol Educ Book. 2025 Jun;45(3): e473082
      The choice of frontline therapy for a patient with chronic phase chronic myeloid leukemia (CP-CML) can have a profound effect on the long-term clinical outcome. Currently, five tyrosine kinase inhibitors (TKIs-imatinib, dasatinib, nilotinib, bosutinib, and asciminib) are available for frontline therapy, but no single TKI is optimal for all patients. EUTOS long-term survival (ELTS) risk score, comorbidities, and treatment-free remission (TFR) priority are the key determinants of frontline TKI selection. Higher ELTS score, low age and comorbidity score, and a high priority for achievement of TFR would all favor the frontline use of a more potent TKI than imatinib. However, no TKI has improved survival compared with imatinib. In children with CP-CML, imatinib, dasatinib, and nilotinib have similar long-term efficacy, with ease of administration and impact of toxicities on quality of life being key considerations. Recent adult trials of reduced-dose dasatinib frontline showed that efficacy may be equivalent to standard-dose dasatinib with a better tolerability and safety profile, but experience is limited in patients with high-risk ELTS scores. The ASC4FIRST trial has confirmed that tolerability and molecular response with asciminib are superior to those with both imatinib and the second-generation (2G)-TKIs. While the overall treatment failure rate was lower with asciminib, the rate of BCR::ABL1 mutations that emerged with asciminib appeared to be higher. The risk of emergent mutations appears to be highly associated with the presence of ASXL1 mutations in the CML cells at diagnosis, but more work is needed to understand the implications of this finding.
    DOI:  https://doi.org/10.1200/EDBK-25-473082
  3. Blood Cancer J. 2025 Apr 23. 15(1): 73
      Autologous stem cell transplantation (ASCT) has been the prime consolidative strategy to increase the depth and duration of response in newly diagnosed multiple myeloma (NDMM), albeit with short- and long-term toxicities. Minimal residual disease (MRD) is an important early response endpoint correlating with clinically meaningful outcomes and may be used to isolate the effect of ASCT. We report the impact of ASCT on MRD burden and generate a benchmark for evaluation of novel treatments as consolidation. We collected MRD by next generation sequencing (NGS; clonoSEQ®) post induction and post-ASCT in consecutive patients (N = 330, quadruplet, N = 279; triplet, N = 51). For patients receiving quadruplets, MRD < 10-5 post-induction was 29% (MRD < 10-6 15%) increasing to 59% post-ASCT (MRD < 10-6 45%). Among patients with MRD > 10-5 post-induction, ASCT lowered the MRD burden>1 log10 for 69% patients. The use of quadruplet induction (vs. triplet) did not reduce the effect of ASCT on MRD burden. Reduction in MRD burden with ASCT was most pronounced in patients with high-risk chromosome abnormalities.This dataset provides granular data to delineate the impact of ASCT on MRD as legacy consolidative strategy in NDMM and provides an important benchmark for evaluation of efficacy of TCRT as experimental consolidative strategy.
    DOI:  https://doi.org/10.1038/s41408-025-01252-6
  4. Front Immunol. 2025 ;16 1558275
      Chimeric antigen receptor (CAR) cellular therapies have advanced outcomes in challenging hematologic malignancies like leukemia, lymphoma, and multiple myeloma. Plasma cell-directed CAR T-cell therapies have been particularly beneficial in multiple myeloma, suggesting that these agents may have a role in other challenging plasma cell disorders such as systemic AL amyloidosis and plasma cell leukemia. AL amyloidosis is a monoclonal plasma cell disorder resulting in the deposition of protein fibrils that compromise end-organ function. Delays in diagnosis can result in end-organ dysfunction and organ failure, making designing and completing treatment difficult. Plasma cell leukemia (PCL) is a rare and highly challenging malignancy with dismal survival outcomes despite aggressive therapy. Both diagnoses are currently treated with regimens borrowed from myeloma: a combination of novel agents and chemotherapy induction, then autologous stem cell transplantation (ASCT), with the current practice trending towards consolidation and maintenance. Unfortunately, only 20% of AL amyloidosis patients are transplant-eligible at diagnosis. Those transplant-ineligible (TIE) patients are treated with combination induction chemotherapy, which may be limited by worsening disease-related end-organ dysfunction. Plasma cell leukemia patients are still very likely to relapse after this intensive and prolonged therapy. Despite the promise of a shorter course of therapy, CAR T-cell therapies directed against plasma cells have not been rigorously investigated in patients with AL amyloidosis or PCL; most trials of MM have excluded these patients. Herein, we describe current treatment paradigms for AL amyloidosis and PCL and review the evidence for CAR T-cell therapies in these challenging plasma cell disorders. Further investigation into CAR T-cell therapies for plasma cell disorders other than multiple myeloma is warranted.
    Keywords:  AL amyloidosis; CAR T-cell; immunotherapy; multiple myeloma; plasma cell dyscrasia; plasma cell leukemia; systemic light chain amyloidosis
    DOI:  https://doi.org/10.3389/fimmu.2025.1558275
  5. Blood Res. 2025 Apr 22. 60(1): 27
       PURPOSE: Primary mediastinal large B-cell lymphoma (PMBCL) is a rare subtype of diffuse large B-cell lymphoma. Radiation therapy (RT) has served as the primary treatment option for PMBCL; however, its role has been questioned with the advent of intensified immunochemotherapy. This study aimed to investigate the role of consolidative RT in the primary treatment of PMBCL.
    METHODS: This single-center retrospective study analyzed the survival outcomes of 65 patients newly diagnosed with PMBCL. The patients were divided into three treatment groups: (1) EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab), (2) R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone), and (3) R-CHOP with consolidative RT.
    RESULTS: The objective response and complete remission rates were 86.2% and 63.1%, respectively, with 3-year progression-free survival (PFS) and overall survival (OS) rates of 72% and 81%, respectively. All patients in the R-CHOP + RT group achieved an objective response with better PFS) than those who did not receive consolidative RT (p = 0.028), although there was no significant difference in OS (p = 0.102). Consolidative RT benefited patients with an initially bulky disease or insufficient end-of-treatment response. The predictive value of 18F-fluorodeoxyglucose positron-emission tomography-computed tomography (PET-CT) in assessing the treatment response in PMBCL was revalidated, showing that patients who achieved negative end-of-treatment PET-CT had significantly better survival outcomes than others.
    CONCLUSIONS: R-CHOP is a useful alternative regimen when intensified chemotherapy is not feasible. Consolidative RT should be considered in cases with an initially bulky disease and insufficient end-of-treatment response.
    Keywords:  Consolidative treatment; Front-line immunochemotherapy; Primary mediastinal large B-cell lymphoma; Radiation therapy
    DOI:  https://doi.org/10.1007/s44313-025-00076-4
  6. Ann Hematol. 2025 Apr 25.
      To characterise the effectiveness of Bruton tyrosine kinase inhibitors with venetoclax in patients with refractory or relapsed mantle cell lymphoma, with or without the addition of an anti-CD20 antibody. Progression-free and overall survival were estimated for forty-nine patients treated with Bruton tyrosine kinase inhibitors and venetoclax (200 mg, daily) or in combination with an anti-CD20 monoclonal antibody between June 2018 and February 2022 in China. The median number of treatment lines before combination therapy was three (range, 2-7). The median patient age was 62 years, with a male-to-female ratio of 3.08:1. Patients exhibited high-risk features including Ki-67 ≥ 30% (89.8%), blastoid/pleomorphic histology (36.7%), high-risk mantle cell lymphoma International Prognostic Index group (42.9%), complex karyotype (27.7%), TP53 mutations (71.4%), TP53 mutations combined with other high-risk gene mutations including KMT2D, NSD2, CCND1, NOTCH1, CDKN2A, NOTCH2 and SMARCA4 (57.1%), and progression of disease within 24 months (65.3%), with similar efficacy and prognosis to low-risk cases. Basic clinical and cytogenetic characteristics, as well as efficacy and survival, were similar between the dual and triple combination therapy groups (all p > 0.05). The optimal overall response and complete remission rates were 67.4% and 53.1%, respectively. The 3-year progression-free and overall survival rates were 37.5% and 50.8%, respectively. Eastern Cooperative Oncology Group≥2was an independent predictor of progression-free survival. Eastern Cooperative Oncology Group performance status ≥ 2, TP53 mutations combined with other high-risk gene mutationswere independent factors for poor overall survival. The most common adverse reactions were haematological and pulmonary infections. The leading cause of death was disease progression (19/22). The combination of Bruton tyrosine kinase inhibitors and venetoclax, demonstrated good efficacy in patients with refractory or relapsed mantle cell lymphoma, particularly in the early treatment. There was no efficacy or survival advantages of adding CD20 antibodies. Patients in the ultrahigh-risk group required more aggressive treatments.
    Keywords:  Combination therapy; High-risk mantle cell lymphoma; Relapse/refractory mantle cell lymphoma
    DOI:  https://doi.org/10.1007/s00277-025-06379-x
  7. Expert Rev Anticancer Ther. 2025 Apr 19.
       INTRODUCTION: The treatment landscape for relapsed refractory (RR) follicular lymphoma (FL) is rapidly evolving, and chemo-free immunotherapeutic strategies represent the major players in the field. CD20×CD3 bispecific antibodies are effective and easily manageable agents that are able to redirect the cytotoxic activity of T-cells against lymphoma cells by creating an immunological synapse.
    AREAS COVERED: In this paper we will review the efficacy and safety profile of the bispecific antibody epcoritamab in RR FL, either as monotherapy (as it was recently approved in United States and Europe) and in combination with other agents. We will discuss its potential role among the existing treatment options in this setting, particularly in relation with other approved bispecific antibodies and with CAR-T cell therapy. Presented data were obtained by literature search on Pubmed and updated with most recent evidence presented at international hematology meetings (2022-2024).
    EXPERT OPINION: the optimal treatment sequencing in FL is still an open issue, however epcoritamab effectively combines key aspects such as high, rapid and durable responses, good tolerability, predictable cytokine release syndrome (CRS) kinetics, and ease of administration, representing an appealing option for patients and clinicians. Future efforts should aim at positioning epcoritamab-based treatments in earlier lines of FL.
    Keywords:  Bispecific antibodies; epcoritamab; follicular lymphoma; immunotherapy; lymphoma
    DOI:  https://doi.org/10.1080/14737140.2025.2492787
  8. J Hematol Oncol. 2025 Apr 24. 18(1): 48
      High-dose chemotherapy followed by autologous hematopoietic stem cell transplantation has significantly improved overall survival (OS) in primary central nervous system lymphoma (PCNSL). However, early identification of long-term survivors remains a challenge. Progression-free survival at 24 months (PFS24) has emerged as a key prognostic marker in diffuse large B-cell lymphoma, but its relevance in PCNSL is still unclear. In this retrospective multicenter study, we analyzed data from 146 newly diagnosed, transplant-eligible PCNSL patients treated with MATRix-like regimens across 14 hospitals. With a median follow-up of 48 months, the 2-year PFS and OS rates were 50.4% and 65.6%, respectively. Of the 139 patients evaluable for PFS24-analysis, 51.1% reached PFS24, with a subsequent 5-year OS of 96.7%. Of note, the annual hazard rate for progression and death decreased to under 5% after 24 months, remaining stable thereafter. The patients who failed to reach PFS24 had a median OS of only 6.0 months. Key predictors of PFS failure included impaired Karnofsky performance status and treatment dose-reduction. In conclusion, PFS24 was identified as an important prognostic marker in PCNSL. Patients who achieve PFS24 have a favorable prognosis, whereas those who do not face poor outcomes and require innovative treatment approaches. This insight could aid in risk stratification and support the use of PFS24 as a surrogate endpoint in clinical trials.
    Keywords:  Overall survival; PFS24; Primary CNS lymphoma
    DOI:  https://doi.org/10.1186/s13045-025-01700-7
  9. Br J Haematol. 2025 Apr 22.
      Multiple myeloma (MM) with chromosome 1q21 gain/amplification (1q+) has been reported to respond poorly to daratumumab. We aimed to explore the mechanism of daratumumab resistance in 1q+ MM. Our findings revealed significantly lower CD38 expression in patients with 1q+ MM than in those with 1q wild type (WT) MM. Next, we focused on the interleukin-6 receptor (IL6R) located in the 1q21 region because a previous report shows that interleukin-6 (IL-6) reduces CD38 expression via the IL-6/Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway activation in MM. Indeed, IL6R expression was significantly higher in 1q+ MM than in 1q WT MM. We verified that the 1q+ human myeloma cell lines (HMCLs) expressed higher IL6R levels than the 1q WT HMCLs. IL-6 treatment induced CD38 downregulation in both the 1q+ HMCLs and primary bone marrow (BM) samples but not in their 1q WT HMCLs and BM samples. IL-6 also resulted in the upregulation of phosphorylated STAT3 in 1q+ HMCLs but not in the 1q WT HMCLs. Furthermore, inhibition of the IL-6/JAK/STAT pathway by treatment with ruxolitinib or tocilizumab restored CD38 expression in the 1q+ HMCLs and BM samples. These findings elucidate the mechanisms underlying daratumumab resistance in 1q+ MM and provide insights for future therapeutic strategies.
    Keywords:  CD38; IL‐6/JAK/STAT pathway; chromosome 1q gain; daratumumab; multiple myeloma
    DOI:  https://doi.org/10.1111/bjh.20106
  10. Hemasphere. 2025 Apr;9(4): e70127
      Patients with newly-diagnosed multiple myeloma (MM) who experience early relapse (ER) have dismal overall survival (OS). Their prospective identification, either before or soon after treatment initiation, is paramount to use alternative approaches and prevent ER. In this study, we investigated the frequency and disease characteristics of ER during the first 18 months after treatment initiation (ER18), in a series of 1215 newly-diagnosed MM patients enrolled in four PETHEMA/GEM clinical trials for the transplant-eligible and transplant-ineligible populations. ER18 was observed in 266 of the 1215 patients (22%) and resulted in a median OS of 19 versus 114 months in cases without ER18. When compared to the ISS and the presence of ≥2 high-risk cytogenetic abnormalities, a modified version of the new high-risk definition from the International Myeloma Society (mHR-IMS) showed the most balanced negative and positive predictive values of ER18 (83.5% and 40%, respectively). In addition to the mHR-IMS, an ECOG = 2, ISS 3, and calcium levels ≥ 11 mg/dL were independently associated with ER18. These variables were modeled into a predictive score in which the rates of ER18 were 2%, 24.5%, and 59% in patients with low-, intermediate-, and high-risk score. The risk of ER18 and OS were modulated by the VGPR status at 6-9 months after treatment initiation. In conclusion, we present a risk model that predicts ER18 and can be readily applied in clinical trials and routine practice to identify treatment strategies empowered to prevent ER18 and improve survival outcomes of newly-diagnosed patients with functional high-risk MM.
    DOI:  https://doi.org/10.1002/hem3.70127
  11. Leukemia. 2025 Apr 19.
      Myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) overlap syndromes are a rare group of biologically and clinically connected hematologic malignancies that includes chronic myelomonocytic leukemia (CMML), the most common subtype, as well as atypical chronic myeloid leukemia, MDS/MPN with SF3B1 and thrombocytosis, and MDS/MPN, not otherwise specified. Given their rarity and overlapping clinical features, accurate diagnosis and risk stratification presents a significant challenge. Therapeutic approaches are largely borrowed from either MDS or MPN and the only curative option for appropriate patients is allogeneic stem cell transplantation. Recent advances have started to uncover the pathobiologic basis for these diseases, leading to novel clinical trials for MDS/MPN overlap syndromes, in particular CMML. This review is a practical guide for the diagnosis and management of MDS/MPN overlap syndromes and presents novel therapeutics being specifically designed for these diseases to improve their historically poor outcomes.
    DOI:  https://doi.org/10.1038/s41375-025-02620-8
  12. Hum Genet. 2025 May;144(5): 481-503
      Next-generation sequencing (NGS) has emerged as a powerful tool for advancing research in chronic myeloid leukemia (CML) by providing a deeper understanding of its genetic complexity. Beyond detecting the hallmark BCR::ABL1 fusion gene, NGS has enabled the identification of additional mutations associated with disease progression, therapy resistance, and clonal evolution. NGS also facilitates the detection of rare BCR::ABL1 fusion variants and cryptic rearrangements, offering a more refined genetic characterization of the disease. Additionally, it enhances the study of minimal residual disease (MRD) and evolving resistance patterns, which are crucial for developing targeted therapeutic strategies. However, challenges such as data interpretation, standardization, and cost constraints continue to limit the widespread application of NGS in routine research and clinical settings. This review explores the contributions of NGS to CML research, highlighting its role in uncovering novel genetic alterations, tracking clonal evolution, and identifying potential therapeutic targets. As sequencing technologies evolve, NGS is expected to further shape the future of CML research, providing critical insights that may ultimately refine disease management strategies.
    DOI:  https://doi.org/10.1007/s00439-025-02745-x