Wiley

American Journal of Hematology

Published by Wiley

Online ISSN: 1096-8652

·

Print ISSN: 0361-8609

Disciplines: Hematology

Journal websiteAuthor guidelines

Top-read articles

198 reads in the past 30 days

Practical diagnostic algorithm for myeloproliferative neoplasms.
Morphologic and laboratory features of essential thrombocythemia and other causes of thrombocytosis, especially prefibrotic myelofibrosis. Dacrocytes, “teardrop” cells; LDH, lactate dehydrogenase; leukoerthyroblastosis, presence of immature myeloid cells and nucleated red cells; MCV, mean corpuscular volume; RDW, red cell distribution width. Source: Adapted from Tefferi and Pardanani.⁵⁰
Triple A (AAA) survival model in essential thrombocythemia. Overall survival data among 598 Mayo Clinic patients stratified by Age, Absolute neutrophil count, and Absolute lymphocyte count, median follow‐up 8.4 years.
Thrombosis risk stratification in essential thrombocythemia (ET). Source: Adapted from Barbui et al.⁶⁵
Current treatment algorithm for essential thrombocythemia. *Second‐line treatment in hydroxyurea intolerant or refractory patients is pegylated IFN‐α or busulfan.
Essential thrombocythemia: 2024 update on diagnosis, risk stratification, and management

January 2024

·

1,601 Reads

·

32 Citations

Ayalew Tefferi

·

Alessandro Maria Vannucchi

·

Tiziano Barbui
View access options

Aims and scope


The American Journal of Hematology is devoted to the coverage of blood diseases in humans and in animal models of human disease. We publish research on non-malignant and malignant hematological diseases, including clinical and basic studies in leukemia, hemostasis and thrombosis, immunology, blood banking, and stem cell biology. With fast turnaround times and personalized feedback from experts in the field, we provide a friendly submissions process that works collaboratively across hematology so every paper has a home.

Recent articles


Foci (a. H/E, 400×) of accumulation of Gaucher‐type histiocytes, with scattered iron‐laden cells (asterisk), alternated with foci (b. H/E, 400×) of sclerosis, with admixed histiocytes, small lymphocytes, eosinophilic granulocytes and scattered, CD30‐positive (b. inset) Hodgkin and Reed–Sternberg cells (arrow). PD‐L1 expression (c. PD‐L1, 100×) is differentially expressed among foci of Gaucher disease (dim intensity) and classical Hodgkin lymphoma (high intensity).
A Case of Hodgkin Lymphoma in a Gaucher Disease Patient: Distinguishing Gaucher and Pseudo‐Gaucher Cells
  • Article
  • Publisher preview available

January 2025

Natalia Scaramellini

·

Marta Canzi

·

Elena Cassinerio

·

[...]

·

Irene Motta


Major bleedings and fatal PE at 15 days according to initial presentation of VTE and baseline platelet count.
Impact of Baseline Thrombocytopenia on Early Outcomes in Patients With Acute Venous Thromboembolism

January 2025

·

3 Reads

Managing acute venous thromboembolism (VTE) in patients with thrombocytopenia is challenging. We used data from the RIETE registry to investigate the impact of baseline thrombocytopenia on early VTE‐related outcomes, depending on the initial presentation as pulmonary embolism (PE) or isolated lower‐limb deep vein thrombosis (DVT). From March 2003 to November 2022, 90 418 patients with VTE were included. Thrombocytopenia was categorized as severe (< 50 000/μL, n = 303) or moderate (50 000–99 999/μL, n = 1882). The primary outcome, fatal PE within 15 days after diagnosis, and secondary outcomes, including major bleeding and recurrent VTE, were analyzed using multivariable‐adjusted models. Among 52 703 patients with PE, the 15‐day case‐fatality rates from PE were 5.8% for severe thrombocytopenia, 4.5% for moderate thrombocytopenia, and 1.1% for normal platelet counts. In 37 715 patients with isolated DVT, the cumulative incidence of fatal PE were 0, 0.2%, and 0.05%, respectively. Multivariable analysis revealed a five‐fold increase in the risk for fatal PE in severe thrombocytopenia (adjusted HR: 4.89; 95%CI: 2.55–9.39) without significant differences between severe and moderate thrombocytopenia. Thrombocytopenia, either moderate or severe, was also associated with increased risk for both, major bleeding and recurrent VTE at 15 days. Initial presentation with PE substantially worsened prognosis compared to isolated DVT. In conclusion, in patients with acute VTE, thrombocytopenia at baseline was associated with increased risk of early death from PE, a finding that was driven by the subgroup whose initial presentation was PE.


RKER‐216 epitope in ALK2 overlaps with BMP6 binding site, and the presence of RKER‐216 competitively inhibits BMP6 from binding to ALK2. (A, E) Binding affinity (KD) between ALK2 and RKER‐216 or ALK2 and BMP6 was determined by SPR. RKER‐216 diluted in HBS‐EP+ buffer was injected through a CM4 chip immobilized with ALK2‐Fc at 25 RU (A) while BMP6 diluted in HBS‐EP+ buffer with 30 nM arginine was injected through a CM4 chip immobilized with ALK2‐Fc at 350 RU (E). The results were analyzed by Biacore Insight Evaluation software using 1:1 binding model for RKER‐216 and steady‐state affinity for BMP6. The mean KD ± SEM from 3 separate experiments is reported and a representative sensogram is shown. (B) RKER‐216 binds to regions in ALK2 covering the F2 loop and β4 sheet (blue) as determined by HDX‐MS. The H/D exchange protected regions (blue) were mapped onto a model structure of ALK2 (PDB ID: 7YRU) shown in ribbon (left) and in surface (right) representation in complex with BMP6 structure (PBD ID: 2R52). The model of ALK2:BMP6 complex was generated by aligning individual structure of ALK2 and BMP6 into the structure of ALK1:BMP9 (PDB ID: 4FAO). The sequence alignment between ALK2 and ALK3 are shown (bottom), highlighting the binding region (in blue) of RKER‐216 in ALK2. (C‐D) BLI competition assays were performed in two ways. (C) ALK2 binding site was saturated with 5 nM of RKER‐216 prior to the competition with BMP6 at 0 to 100 nM. In addition to the vehicle control, BMP6 at 100 nM was added to unsaturated ALK2 to confirm binding ability of BMP6 in the competition. No binding activity was observed in the competition, indicating that BMP6 at 100 nM was unable to complete RKER‐216 away for binding ALK2. (D) ALK2 binding site was saturated with 100 nM of BMP6 prior to the competition with RKER‐216 at 0 to 50 nM. RKER‐216 at 50 nM was added to unsaturated ALK2 to confirm binding ability of RKER‐216. This group also served as a control for examining whether RKER‐216 binds to ALK2 or the ALK2‐BMP6 complex, as changes in the wavelength reflects both the size and affinity of the interactor. In the zoomed plot of RKER‐216 competition, data were aligned to zero on the Y‐axis prior to the competition step for visual comparison, and 50 nM RKER‐216 without saturation had the greatest wavelength shift, indicating that RKER‐216 dislodged BMP6 from ALK2 instead of binding to the ALK2‐BMP6 complex. Experiments were repeated 3 times and a representative sensogram is shown.
RKER‐216 decreases hepcidin transcription in vitro and controls iron availability by lowering hepcidin secretion in vivo. (A) Hep3B cells were serum starved with 1% FBS overnight and incubated with an ascending dose of RKER‐216 (0.02–1 μg/mL) in the absence or presence of 5 ng/mL of BMP6, BMP2/6, or BMP2 for 6 h (n = 4–5 per group). (B) Characterization of human ACVR1 knockout in HepG2 and Huh7 cells by quantifying ACVR1 and BMPR1A transcript copy number (n = 3 per group). (C) ACVR1 KO and the negative control cells were treated with RKER‐216 at 1–30 μg/mL for 6 h (n = 3 per group). Relative HAMP mRNA levels were determined by qRT‐PCR and transcripts were normalized to an internal control RPL19. The average of PBS control without ligand stimulation or ACVR1 wildtype without RKER‐216 was set to 1. For ligand preference, hepcidin stimulated by each ligand was set to 100%. Values represent mean ± SEM. Results were compared across RKER‐216 and BMP ligands by one‐ or two‐way ANOVA with Tukey's post hoc test. Means without a common superscript differ significantly (p < 0.05) or *p < 0.05, ***p < 0.001 relative to untreated cells of the same genotype. (D‐G) B6N male mice at 8 weeks were treated with a single SC dose of isotype control or RKER‐216 at 3 mg/kg for times as indicated (n = 5 per group). Serum was collected to quantify (D) RKER‐216 exposure and (E) serum hepcidin by ELISA, and (F) serum iron and (G) serum transferrin saturation (TSAT) by colorimetric assays. Values represent mean ± SEM. *p < 0.05; **p < 0.01; ***p < 0.001 relative to the isotype control mice of the same time point by Student's t‐test.
RKER‐216 inhibits BMP‐SMAD signaling to reduce hepcidin production mediated by inflammation. B6N male mice at 7 weeks were SC‐dosed with isotype control or RKER‐216 at 3 mg/kg for 1 h, followed by an IP injection of PBS or LPS at 1 mg/kg (n = 5 per group). Six hours after LPS administration, livers and serum were analyzed for (A) relative Il6 mRNA and (C) Hamp mRNA by qRT‐PCR, and (B) serum IL‐6 and (D) hepcidin by ELISA. (E) STAT3 phosphorylation and (F) SMAD1/5/9 phosphorylation levels in total liver lysates were determined by immunoblot and normalized to total STAT3 and total SMAD5, respectively. Representative immunoblots are shown. Transcripts were normalized to Rpl19, and the average of PBS treated isotype control mice were set to 1. Values represent mean ± SEM. *p < 0.05, ***p < 0.001 relative to the respective PBS treated control group by two‐way ANOVA with Tukey's post hoc test.
RKER‐216 ameliorates iron‐restricted erythropoiesis by suppressing hepcidin to liberate spleen iron in a mouse model of AI. CKD mice switched to Ade3 diet or continued on Ade40 diet were SC treated with isotype control or RKER‐216 at 3 mg/kg twice weekly for 4 weeks (n = 5–13 per group). Tissues were analyzed for (A) serum hepcidin by ELISA, (B) spleen iron, (C) TSAT and (H) liver iron by colorimetric assays, and (D) hemoglobin, (E) hematocrit, (F) red blood cells (RBC), and (G) reticulocyte hemoglobin by the Element HT5 hematology analyzer. Values represent mean ± SEM. *p < 0.05; **p < 0.01; ***p < 0.001 relative to isotype control mice on the same diet by one‐way ANOVA with Tukey's post hoc test or Welch and Brown–Forsythe ANOVA with Dunnett's post hoc test for those with unequal variances.
RKER‐216 provides the iron necessary to reverse iron‐restricted erythropoiesis and to resolve anemia when coupled with rhEPO in addressing AI. CKD mice on Ade40 diet were dosed with isotype control, RKER‐216 at 3 mg/kg or a combination of RKER‐216 and 3000 U/kg of rhEPO for 4 weeks (n = 15 per group). Age‐ and sex‐matched B6N mice on an AIN‐93G diet were included to establish a healthy baseline (n = 7; non‐CKD). Tissues were analyzed for (A) serum hepcidin by ELISA, (B) spleen iron, (C) TSAT and (H) liver iron by colorimetric assays, (D) hemoglobin, (E) hematocrit, and (F) RBC by the Element HT5 hematology analyzer, and (G) serum EPO by MSD. (I) Spleen and liver sections were stained with Perls' Prussian blue for tissue iron. Images were taken under brightfield, and representative images are shown. Arrows indicate macrophage iron staining in isotype‐treated CKD mouse livers. Values represent mean ± SEM. *p < 0.05, ***p < 0.001 relative to the indicated group by one‐way ANOVA with Tukey's post hoc test or Welch and Brown–Forsythe ANOVA with Dunnett's post hoc test.
A Recombinant Antibody Against ALK2 Promotes Tissue Iron Redistribution and Contributes to Anemia Resolution in a Mouse Model of Anemia of Inflammation

January 2025

·

20 Reads

Patients with chronic inflammation are burdened with anemia of inflammation (AI), where inflammatory cytokines inhibit erythropoiesis, impede erythropoietin production, and limit iron availability by inducing the iron regulator hepcidin. High hepcidin hinders iron absorption and recycling, thereby worsening the impaired erythropoiesis by restricting iron availability. AI management is important as anemia impacts quality of life and potentially affects morbidity and mortality. The bone morphogenetic protein (BMP)‐SMAD pathway is crucial for hepcidin regulation. Here, we characterized a research antibody against BMP receptor ALK2, RKER‐216, and investigated its mechanism in suppressing hepcidin and improving anemia in acute/chronic inflammation. Additive effects of RKER‐216 and recombinant human erythropoietin (rhEPO) on erythropoiesis and iron utilization were also explored. We showed that RKER‐216 neutralized ALK2 activity by competing with the binding of BMP6. RKER‐216 reduced hepcidin transcription in Hep3B cells, and a subcutaneous dose of RKER‐216 at 3 mg/kg suppressed serum hepcidin and increased circulating iron for 3–4 days in wildtype mice. Moreover, RKER‐216 decreased hepcidin by inhibiting SMAD1/5/9 signaling in lipopolysaccharide‐mediated inflammation and liberated iron from the recycling pathway to alleviate anemia in mice with adenine‐induced chronic kidney disease (CKD), a mouse model of AI. Finally, RKER‐216 reversed iron‐restricted erythropoiesis in CKD mice and supplied the iron requirement for complete resolution of anemia when coupled with rhEPO in addressing AI. Our data support that ALK2 is a key hepcidin regulator and that a neutralizing ALK2 antibody has the potential to restore iron homeostasis as monotherapy or in combination with rhEPO to ameliorate AI.


Study selection flowchart.
MRD negativity (OR) (A) and PFS (HR) (B) pooled‐estimates for the whole study population (by MM setting). 95%‐CI, 95% confidence interval; HR, hazard ratio; MM, multiple myeloma; MRD, minimal residual disease; NDMM TE, newly diagnosed multiple myeloma transplant eligible; NDMM TIE, newly diagnosed multiple myeloma transplant ineligible; OR, odds ratio; PFS, progression free survival; RRMM, relapsed/refractory multiple myeloma.
Univariate regression in a meta‐analytic framework for the association of MRD. Negativity (OR) on PFS (HR) (A) and MRD negativity (OR) on OS (HR) (B). 95% CI, 95% confidence interval; HR, hazard ratio; MRD, minimal residual disease; OR, odds ratio; PFS, progression free survival. Regression equation (2A): Log(PFS_HR) = −0.35 − 0.20 × Log(MRDneg_OR), p < 0.001, Regression equation (2B): Log(OS_HR) = −0.12*log(MRDneg_OR), p = 0.023.
Evaluating Minimal Residual Disease Negativity as a Surrogate Endpoint for Treatment Efficacy in Multiple Myeloma: A Meta‐Analysis of Randomized Controlled Trials

January 2025

·

4 Reads

This meta‐analysis examined the association between minimal residual disease (MRD) negativity and survival outcomes in 15 304 patients with multiple myeloma (MM) enrolled in randomized controlled trials published until June 2, 2024. Overall, there was a significant, negative and strong association between MRD negativity odds ratios and survival hazard ratios (β_PFS = ‐0.20, p < 0.001, β_OS = ‐0.12, p = 0.023). These associations remained significant for newly diagnosed patients (β_PFS = ‐0.35, p < 0.001), and they were consistent but not significant for relapsed/refractory patients (β_PFS = ‐0.06, p = 0.635). Sustained MRD negativity at 1 year was strongly correlated with prolonged PFS (β_PFS = ‐0.30, p < 0.001). In conclusion, this comprehensive meta‐analysis supports MRD as a surrogate for survival in MM.


Comparisons of graft‐versus‐host disease and transplant outcomes of haploidentical transplant cohort treated with budesonide vs. 1:1 propensity‐matched CIBMTR controls. (A) Grade II‐IV aGVHD; (B) Grade III‐IV aGVHD; (C) Chronic GVHD; (D) Overall survival; (E) Progression‐free survival; (F) GVHD‐free, relapse‐free survival (GRFS).
GVHD biomarkers: (A) Correlations between aGVHD biomarkers at the time of engraftment and GI symptom onset; (B) aGVHD biomarkers at baseline and GI symptom onset in patients with and without acute GI GVHD; (C) AuROC of intermediate/high‐risk aGVHD biomarker symptom onset panel in predicting NRM within 6 months postsymptom onset.
Budesonide, Added to PTCy‐Based Regimen, for Prevention of Acute GI GVHD After Allogeneic Stem Cell Transplantation

Oral budesonide exerts local effects with negligible systemic glucocorticoid activity, due to rapid first‐pass metabolism, therefore, could potentially be efficacious in preventing gastrointestinal (GI) acute GVHD (aGVHD). We explored the use of budesonide, added to posttransplant cyclophosphamide (PTCy), tacrolimus, and mycophenolate mofetil, for prevention of GI aGVHD after allogeneic hematopoietic stem cell transplantation (AHSCT) in a prospective observational study and treated 80 patients with a median age of 53 years (range 19–74). Results were compared with a publicly available CIBMTR dataset of 646 patients who received PTCy‐based GVHD prophylaxis (CIBMTR Study # GV17‐02) (control). Cumulative incidence (CI) of 3‐month grade 2–4 and grade 3–4 aGVHD in the budesonide and control groups were 3.8% vs. 34.4% (p < 0.001) and 1.3% vs. 9.8% (p = 0.029), respectively. One‐year GRFS (70.5% vs. 31.5%, p < 0.001), PFS (73.4% vs. 52.8%, p = 0.003), and OS (80.1% vs. 64.2%, p = 0.038) were significantly higher in the budesonide group compared with control group. Propensity score‐adjusted analyses showed that the addition of budesonide significantly decreased risk of aGVHD grade 2–4 (HR 0.29, p < 0.001), grade 3–4 (HR 0.12, p = 0.045), and cGVHD (HR 0.22, p < 0.001), which resulted in better GRFS (HR 0.38, p < 0.001), PFS (HR 0.58, p = 0.012), and OS (HR 0.72, p = 0.044). Similar results were found when using propensity score‐matched analysis restricted to recipients of haploidentical transplantation. In conclusion, addition of budesonide to PTCy‐based GVHD prophylaxis is safe and effective in preventing severe acute GI GVHD with significantly improved GRFS. These results could facilitate transition to peripheral blood grafts for all allogeneic transplant recipients.


Overview of molecular markers for detecting MRD in AML. Peripheral blood monitoring of MRD can be subdivided by molecular signatures present in circulating leukemic cells or cell‐free cellular products in the plasma. Cell‐based assays range from interrogating surface proteins with flow cytometry to nucleic acid amplification‐based techniques to identify recurrent DNA/RNA mutations, cytogenetic and epigenetic marks, or overexpressed RNA within circulating tumor cells. Multiple molecular targets are present in PB plasma or packaged within extracellular vesicles (EVs), aiding with the detection of MRD and relapse before circulating blasts are detectable in the blood. This figure was created using BioRender.
Breaking the Bone Marrow Barrier: Peripheral Blood as a Gateway to Measurable Residual Disease Detection in Acute Myelogenous Leukemia

Acute myeloid leukemia (AML) is a genetically heterogeneous disease with high rates of relapse after initial treatment. Identifying measurable residual disease (MRD) following initial therapy is essential to assess response, predict patient outcomes, and identify those in need of additional intervention. Currently, MRD analysis relies on invasive, serial bone marrow (BM) biopsies, which complicate sample availability and processing time and negatively impact patient experience. Additionally, finding a positive result can generate more questions than answers, causing anxiety for both the patient and the provider. Peripheral blood (PB) evaluation has shown promise in detecting MRD and is now recommended by the European Leukemia Net for AML for certain genetic abnormalities. PB‐based sampling allows for more frequent testing intervals and better temporal resolution of malignant expansion while sparing patients additional invasive procedures. In this review, we will discuss the current state of PB testing for MRD evaluation with a focus on next‐generation sequencing methodologies that are capable of MRD detection across AML subtypes.


Flowchart of patient selection.
Reverse Kaplan–Meier plot illustrating time‐to‐relapse detection in patients with relapse detected at routine visits or outside of routine visits in the entire relapse cohort (A), and excluding patients with relapses detected after the pre‐planned routine follow‐up period (B).
Limited Benefit of Routine Clinical Follow‐Up for Relapse Detection in Diffuse Large B‐Cell Lymphoma Patients in Complete Remission Following First‐Line Treatment

Despite advances in treatment, approximately 15% of patients with diffuse large B‐cell lymphoma (DLBCL) who achieve complete remission (CR) after first‐line therapy will experience a relapse. However, there is no consensus on the optimal follow‐up strategies for detecting relapse after achieving CR. This population‐based study, based on the Danish Lymphoma Registry (LYFO), identified a total of 1634 patients diagnosed with DLBCL between 2010 and 2017, including 105 patients who achieved CR following first‐line R‐CHOP‐like therapy and subsequently relapsed. The median follow‐up time was 6 years (range 3–8 years). Most cases of relapse were symptomatic (83%), with B symptoms and peripheral lymphadenopathy being the most common. Asymptomatic relapses were identified through physical examination (1%), blood tests (3%), or imaging findings (13%). The proportion of relapses identified outside routine visits was 70%. Only 5% of scheduled routine visits led to a relapse diagnosis, whereas 74% of unscheduled visits initiated by the patient outside routine follow‐up resulted in relapse detection. Our findings highlight that systematic, scheduled monitoring of patients in remission after first‐line treatment contributes only modestly to the early detection of disease recurrence. Future studies should explore alternative methods of relapse surveillance rather than relying solely on pre‐scheduled clinical follow‐up.


Consort diagram.
(A) Relapse‐free and overall survival for the totality of patients treated in the two cohorts, (B) overall survival by cohort, and (C) relapse‐free survival by cohort.
Hyper‐CVAD and Sequential Blinatumomab Without and With Inotuzumab in Young Adults With Newly Diagnosed Philadelphia Chromosome‐Negative B‐Cell Acute Lymphoblastic Leukemia

Adding inotuzumab ozogamicin (InO) to hyper‐CVAD and blinatumomab may improve outcomes in newly diagnosed Philadelphia chromosome (Ph)‐negative B‐cell acute lymphoblastic leukemia (B‐ALL). Patients with newly diagnosed B‐ALL received up to four cycles of hyper‐CVAD followed by four cycles of blinatumomab. Beginning with patient #39, InO 0.3 mg/m² was added on Days 1 and 8 to two cycles of high‐dose methotrexate and cytarabine, and two cycles of blinatumomab. The primary endpoint was the relapse‐free survival (RFS) rate. Seventy‐five patients were treated (median age of 33 years; range, 18–59), of whom 37 (49%) received hyper‐CVAD with blinatumomab and InO (cohort 2). Measurable residual disease (MRD) negativity by next‐generation sequencing (sensitivity: 1 × 10⁻⁶) was achieved in 79% of patients in cohort 2. The median follow‐up was 44 months (range, 13–90) overall, and 26 months (range, 8–39) in cohort 2. For the entire cohort, the estimated 3‐year RFS rate was 82% and the 3‐year overall survival rate was 90%. These rates were 90% versus 74% (p = 0.06) and 100% versus 82% (p = 0.01) in patients who did or did not receive InO, respectively. No sinusoidal obstruction syndrome was observed. In summary, hyper‐CVAD with blinatumomab and InO improved the outcomes of patients with newly diagnosed B‐ALL.






Response characteristics of study patients. CRc, composite complete response; MLFS, morphological leukemia‐free state; NR, no response. [Color figure can be viewed at wileyonlinelibrary.com]
Survival outcomes of study patients. [Color figure can be viewed at wileyonlinelibrary.com]
Comparative survival outcomes of patients with untreated secondary AML (s‐AML) and treated s‐AML. [Color figure can be viewed at wileyonlinelibrary.com]
Overall survival (OS) of patients treated with venetoclax and stratified by TP53 status and other risk models. IC, intensive chemotherapy; Int, intermediate; LIT, low‐intensity treatment; mos, months; NAR, number at risk; Ven, venetoclax. [Color figure can be viewed at wileyonlinelibrary.com]
Comparison of overall survival (OS) between transplanted patients and a nontransplanted landmark comparator*. NAR, number at risk; SCT, allogeneic hematopoietic stem cell transplantation. *The landmark comparator consisted of patients < 70 years of age at AML diagnosis, with an overall response, and alive for ≥ 3.2 months. [Color figure can be viewed at wileyonlinelibrary.com]
Outcomes of Patients With Treated Secondary Acute Myeloid Leukemia: A High‐Risk Subtype That Warrants an Independent Prognostic Designation

December 2024

·

38 Reads

Patients who develop acute myeloid leukemia (AML) after having received treatment for myelodysplastic syndrome (MDS) or related conditions have particularly poor outcomes. This study analyzed adult patients with newly diagnosed AML who previously had MDS, chronic myelomonocytic leukemia (CMML), or MDS/myeloproliferative neoplasm (MPN) overlap syndrome, and who had received hypomethylating agents, chemotherapy, and/or allogeneic stem cell transplantation (HSCT) for these antecedent disorders. From January 2012 to August 2023, we included 673 patients with a median age of 70 years (range, 19–94); 536 (80%) had transformed from MDS, and the remainder from CMML or MDS‐MPN. Additionally, 149 patients (22%) had prior therapy for nonmyeloid malignancies. Among 497 evaluable patients, 289 (58%) had adverse‐risk (AR) cytogenetics, 34% had TP53 mutation/s, and 71% were classified as AR by the ELN 2017 criteria. Most patients (67%) received low‐intensity therapy (LIT) for AML, and 27% were treated with venetoclax. The overall response rate was 37%, and venetoclax improved the odds of response (OR = 2.5, 95% CI 1.6–3.7) in LIT–treated patients. At a median follow‐up of 43 months, the median relapse‐free survival (RFS) and overall survival (OS) were 4.6 and 4.8 months, respectively. Multivariate analysis showed that prior therapy for nonmyeloid disorders (HR = 1.30), ≥ 2 lines of therapy for antecedent myeloid disorders (HR = 1.23), and ELN AR risk (HR = 1.47) increased the hazards of death, while HSCT (HR = 0.50) was beneficial and validated on gradient‐boosted regression. TS‐AML is associated with poor outcomes irrespective of AML genomics and treatment, highlighting the need for its inclusion as an independent AR category for accurate prognostication and clinical trial reporting.





Duration of response and survival outcomes. (A) Duration of overall response, (B) duration of complete response, (C) progression/relapse‐free survival, (D) overall survival, (E) cumulative incidence of non‐relapse/progression mortality (NRM). [Color figure can be viewed at wileyonlinelibrary.com]
Outcomes of CD19 CAR T in Transformed Indolent Lymphoma Compared to De Novo Aggressive Large B‐Cell Lymphoma

December 2024

·

6 Reads

Chimeric antigen receptor (CAR) T‐cell therapy has revolutionized treatment of aggressive large B‐cell lymphoma (aLBCL). Patients with transformed indolent non‐Hodgkin lymphoma (tiNHL) were included in key CAR trials, but outcomes of CAR for this distinct, historically high‐risk group are poorly understood. We conducted a multicenter retrospective study of 1182 patients with aLBCL receiving standard‐of‐care CAR T between 2017 and 2022, including 338 (29%) with tiNHL. Rates of grade ≥ 3 cytokine release syndrome (CRS) were similar between tiNHL and de novo cohorts (7% vs. 8%, p = 0.6), while grade ≥ 3 immune effector cell‐associated neurotoxicity syndrome was lower in tiNHL (21% vs. 27%, p = 0.02). Overall response rate was similar in both cohorts (83% vs. 81%, p = 0.3), while complete response rate was higher in tiNHL (67% vs. 59%, p = 0.017). With a median follow‐up of 22.3 months, the progression/relapse‐free (PFS) and overall survival (OS) were similar between the tiNHL and de novo cohorts (24‐month PFS 41% [95% CI: 35%–46%] vs. 38% [95% CI: 35%–42%]; 24‐month OS 58% [95% CI: 52%–63%] vs. 52% [95% CI: 48%–56%], respectively). After adjusting for key risk factors, there was a trend toward a lower hazard of disease progression, relapse or death post‐CAR for tiNHL patients compared to de novo aLBCL patients (HR: 0.84 [95% CI: 0.69–1.0], p = 0.07). Elevated LDH, advanced stage, prior bendamustine within 12 months of CAR, receipt of bridging therapy, CNS involvement, and ≥ 3 prior lines of therapy were each associated with inferior PFS. In conclusion, CAR T therapy is highly effective with an acceptable toxicity profile in patients with tiNHL.


Community‐acquired respiratory viral infection distribution per baseline hematological malignancy in EPICOVIDEHA‐EPIFLUEHA participants with hematological patients diagnosed with respiratory viral infections (September 2023–March 2024).
Day 30 survival probability by respiratory viral pathogen.
Respiratory Viruses in Patients With Hematological Malignancy in Boreal Autumn/Winter 2023–2024: EPICOVIDEHA‐EPIFLUEHA Report

December 2024

·

132 Reads

Community‐acquired respiratory viral infections (CARV) significantly impact patients with hematological malignancies (HM), leading to high morbidity and mortality. However, large‐scale, real‐world data on CARV in these patients is limited. This study analyzed data from the EPICOVIDEHA‐EPIFLUEHA registry, focusing on patients with HM diagnosed with CARV during the 2023–2024 autumn–winter season. The study assessed epidemiology, clinical characteristics, risk factors, and outcomes. The study examined 1312 patients with HM diagnosed with CARV during the 2023–2024 autumn–winter season. Of these, 59.5% required hospitalization, with 13.5% needing ICU admission. The overall mortality rate was 10.6%, varying by virus: parainfluenza (21.3%), influenza (8.8%), metapneumovirus (7.1%), RSV (5.9%), or SARS‐CoV‐2 (5.0%). Poor outcomes were significantly associated with smoking history, severe lymphopenia, secondary bacterial infections, and ICU admission. This study highlights the severe risk CARV poses to patients with HM, especially those undergoing active treatment. The high rates of hospitalization and mortality stress the need for better prevention, early diagnosis, and targeted therapies. Given the severe outcomes with certain viruses like parainfluenza, tailored strategies are crucial to improving patient outcomes in future CARV seasons.









Journal metrics


10.1 (2023)

Journal Impact Factor™


21%

Acceptance rate


15.7 (2023)

CiteScore™


4 days

Submission to first decision


$5,190 / £3,460 / €4,330

Article processing charge

Editors