{"title":"MECHANISMS OF RESISTANCE TO T-CELL REDIRECTING THERAPIES","authors":"A. A. Alizadeh","doi":"10.1002/hon.70093_68","DOIUrl":"https://doi.org/10.1002/hon.70093_68","url":null,"abstract":"<p>T-cell redirecting therapies, have revolutionized the management of diverse malignancies, especially B-cell non-Hodgkin lymphomas. Currently approved such therapies include T-cells that are engineered to express chimeric antigen receptors (CAR-T cells) and bispecific antibodies that bridge T-cells to diverse tumor antigens. Yet despite their remarkable efficacy and curative potential, resistance and relapse to these therapies remain significant hurdles, and unfortunately, still observed in most patients today. This presentation will explore the tumor-intrinsic mechanisms that contribute to such resistance in mature B-cell neoplasms, while also considering the roles of T-cell dysfunction and the tumor microenvironment (TME) in resistance phenotypes.</p><p>I will review data from several studies that have highlighted key mechanisms and pathways underlying such tumor intrinsic resistance. In one key study by Sworder et al. (<i>Cancer Cell</i>, 2023) a comprehensive simultaneous tumor and effector profiling (STEP) approach has been described to investigate resistance determinants in large B-cell lymphomas treated with anti-CD19 CAR T-cells. In addition to genetic and epigenetic mechanisms known to hamper target antigen expression of key tumor markers (such as CD19, CD20, CD22, and BCMA), these studies have revealed that genetic alterations in B cell identity genes like PAX5 and IRF8 may lead to lineage switch or loss of target antigens. Separately, these studies show how somatic gains driving upregulation of key immune checkpoints like PD-L1 upregulation can help tumors evade T-cell attacks. For bispecific antibodies, similar mechanisms, such as antigen loss, are also observed, suggesting shared challenges across therapies.</p><p>When considering non-tumor intrinsic mechanisms of resistance, the TME is also known to play a crucial role, with research indicating that immune-suppressed TME profiles correlate with poorer outcomes, likely by hindering T-cell function through axes such as PD-1, TIM-3, suppressive actions of Tregs, MDSCs, inhibitory cytokines, and others. T-cell exhaustion, driven by chronic antigen exposure and TME immunosuppression, is known to reduce effector functions and persistence, impacting both CAR-T and bispecific antibody efficacy. Conversely, TMEs with high B cell proliferation may predict better CAR-T responses, an unexpected feature that could also guide therapy selection.</p><p>I will highlight how such approaches to integrative genomic profiling, TME analysis, and T-cell functional assessments can enhance outcome prediction and personalize T-cell therapies. In addition to defining key gaps in our current knowledge, I will describe strategies to help bridge these gaps, toward optimizing existing therapies and developing next-generation interventions to overcome resistance, potentially improving long-term outcomes for patients with lymphomas and other tumors.</p><p><b>Keywords:</b> aggressive B-cell non-Hodgkin lymphoma</p","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70093_68","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292218","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}
J. S. Abramson, M. Ku, M. Hertzberg, C. P. Fox, C. Herbaux, H. Huang, D. H. Yoon, W. S. Kim, H. Zhang, H. Abdulhaq, W. Townsend, E. Mulvihill, V. Orellana-Noia, R. Ta, H. Huang, M. J. Kallemeijn, A. Belousov, A. Bottos, L. Lundberg, G. P. Gregory
{"title":"GLOFITAMAB PLUS GEMCITABINE AND OXALIPLATIN (Glofit-GemOx) IN PATIENTS WITH RELAPSED/REFRACTORY (R/R) DIFFUSE LARGE B-CELL LYMPHOMA (DLBCL): 2-YEAR FOLLOW-UP OF STARGLO","authors":"J. S. Abramson, M. Ku, M. Hertzberg, C. P. Fox, C. Herbaux, H. Huang, D. H. Yoon, W. S. Kim, H. Zhang, H. Abdulhaq, W. Townsend, E. Mulvihill, V. Orellana-Noia, R. Ta, H. Huang, M. J. Kallemeijn, A. Belousov, A. Bottos, L. Lundberg, G. P. Gregory","doi":"10.1002/hon.70093_76","DOIUrl":"https://doi.org/10.1002/hon.70093_76","url":null,"abstract":"<p><b>Introduction:</b> Glofitamab, a CD20:CD3 bispecific antibody, has shown durable responses as fixed duration monotherapy in R/R DLBCL after ≥ 2 prior lines of therapy (LOT; Dickinson et al. NEJM 2022). In the Phase 3 STARGLO trial, Glofit-GemOx demonstrated overall survival (OS) and progression-free survival (PFS) benefits over rituximab (R)-GemOx in autologous stem cell transplant (ASCT)-ineligible R/R DLBCL (Abramson et al. Lancet 2024). Here, we present updated efficacy and safety from the STARGLO trial (NCT04408638), including landmark analyses of patients (pts) in complete remission (CR).</p><p><b>Methods:</b> Pts were randomized 2:1 to receive Glofit-GemOx (8 cycles plus 4 cycles glofitamab monotherapy) or R-GemOx (8 cycles) and stratified by number of prior LOT (1 vs. ≥ 2) and refractoriness to their last therapy. After obinutuzumab pretreatment, glofitamab was given in Cycle (C) 1 as weekly step-up doses (2.5/10 mg) then 30 mg target dose every 21 days from C2 Day 1. Pts with only 1 prior LOT must have been ASCT-ineligible. Primary endpoint was OS. Secondary endpoints included independent review committee (IRC)-assessed PFS and CR rate. A landmark analysis of pts in CR at end of treatment (EOT) was performed.</p><p><b>Results:</b> Of 274 pts (Glofit-GemOx, <i>n</i> = 183; R-GemOx, <i>n</i> = 91), 172 (62.8%) had 1 prior LOT, 102 (37.2%) had ≥ 2 prior LOT, 153 (55.8%) were primary refractory, and 166 (60.6%) were refractory to their last therapy. Baseline characteristics were unchanged compared with the primary analysis and well balanced across arms.</p><p>With 2 years (yrs) of follow-up (data cut off: June 17, 2024; median follow-up: 24.7 months [mo]), Glofit-GemOx continued to confer superior OS (median: not evaluable [NE] vs. 13.5 mo; hazard ratio [HR] 0.60, 95% confidence interval [CI]: 0.42–0.85), median IRC-assessed PFS (13.8 vs. 3.6 mo; HR 0.41, 95% CI: 0.29–0.58), and CR rate (58.5 vs. 25.3%) versus R-GemOx. For Glofit-GemOx-treated pts in CR (<i>n</i> = 107), median duration of CR was not reached (95% CI: 27.2–NE; median CR follow-up, 18.2 mo [range: 15.2–19.3]). In pts with a CR at EOT (<i>n</i> = 82), the OS and PFS rates 1 yr after EOT were 89.3% and 82.4%, respectively.</p><p>The Glofit-GemOx safety profile was unchanged. Cytokine release syndrome (CRS) was the most common adverse event in glofitamab-exposed pts (Grade [Gr] 1, 32.0%; Gr 2, 10.5%; Gr 3, 2.3%). Events consistent with immune effector cell-associated neurotoxicity syndrome occurred in 4 pts (all concurrent with CRS; most Gr 1–2 [<i>n</i> = 3]). Exploratory biomarker and immune recovery data will be presented.</p><p><b>Conclusions:</b> With 2 yrs of follow-up, Glofit-GemOx sustained a clinically meaningful benefit in OS and PFS versus R-GemOx in ASCT-ineligible pts with R/R DLBCL, with most (82%) pts in CR at EOT still in remission. The safety profile was consistent with known risks of each drug. The updated analyses demonstrate durable remissions and maintain","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70093_76","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292221","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}
C. Querfeld, L. Chen, X. Wu, Z. Han, C. Su, Y. Yuan, M. Banez, J. Quach, T. Barnhizer, L. Crisan, S. T. Rosen, J. Zain
{"title":"DURVALUMAB AND LENALIDOMIDE SHOWS SUPERIOR EFFICACY OVER SINGLE-AGENT DURVALUMAB IN REFRACTORY/ADVANCED CUTANEOUS T CELL LYMPHOMA: RESULTS FROM A RANDOMIZED PHASE 2 TRIAL","authors":"C. Querfeld, L. Chen, X. Wu, Z. Han, C. Su, Y. Yuan, M. Banez, J. Quach, T. Barnhizer, L. Crisan, S. T. Rosen, J. Zain","doi":"10.1002/hon.70094_399","DOIUrl":"https://doi.org/10.1002/hon.70094_399","url":null,"abstract":"<p><b>Introduction:</b> Advanced stages of cutaneous T cell lymphoma (CTCL) have an unfavorable prognosis. We have shown that CTCL escapes immune surveillance via immune checkpoint signaling such as the PD-1/PD-L1 axis. Selective targeting of the functionally exhausted malignant T cells in cutaneous T-cell lymphoma (CTCL) and distinct cells within the tumor microenvironment (TME) via PD1/PD-L1 blockade (durvalumab) may restore an anti-tumor immune response. We initiated the randomized Phase 2 portion to compare single agent durvalumab to durvalumab plus lenalidomide in relapsed/advanced CTCL (NCT03011814). The primary end point was objective response rate (ORR) using the global composite response (based on skin, blood, nodes, and viscera) according to consensus guidelines. Secondary end points included duration of response, progression-free survival, and toxicity. Relationships between gene expression profile, tumor-microenvironment (TME), and antitumor activity were exploratory end points.</p><p><b>Methods:</b> Adult patients with histologically confirmed MF or SS, who had failed ≥ 2 systemic therapies were enrolled and randomized 1:1 to single agent durvalumab (1500 mg (day 1 of 28-day cycle) or durvalumab (same dose) & lenalidomide (10 mg for cycle 1, 15 mg for cycle 2, then 20 mg for subsequent cycles daily for 21 days of each 28-day cycle). The study used a “pick a winner” design based on ORR. Serial skin and blood samples were collected to assess the impact on the TME and anti-tumor activity.</p><p><b>Results:</b> Among 25 patients [12 durvalumab; 13 durvalumab/lenalidomide; stage IB, 2 (17%) vs. 4 (31%); stage IIB, 5 (42%) vs. 3 (23%); stage III/IV, 5 (42%) vs. 6 (46%); large cell transformation 3 (25%) vs. 5 (38%)], the combination showed superior clinical activity, with an ORR of 75% versus 42% and a 12-month PFS of 73% (95% CI: 38%–91%) versus 36% (95% CI: 11%–63%) (Figure 1). Median PFS was 6.2 months for durvalumab and not reached for the combination. The most common treatment-emergent adverse events were more frequent in the durvalumab/lenalidomide arm versus durvalumab arm and included fatigue (<i>n</i> = 10), diarrhea (<i>n</i> = 6), anemia (5), decreased platelets (<i>n</i> = 5), leukopenia & neutropenia (<i>n</i> = 4), constipation (<i>n</i> = 4), and leg edema (<i>n</i> = 4). The majority of AEs with both treatment arms were mild to moderate in severity (grade I/II, 92%; grade III, 8 %). One grade IV neutropenia on combo arm was observed.</p><p><b>Conclusions:</b> This randomized phase 2 trial of durvalumab +/− lenalidomide evaluating anti-tumor activity demonstrated superior clinical activity of combinatorial durvalumab/lenalidomide versus single-agent durvalumab in refractory/advanced CTCL. Responses were durable and ongoing, and treatment was well tolerated. Our correlative results from sequential skin biopsies demonstrated immune signatures for enhanced anti-tumor responses that may be predictive of response to check","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_399","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292473","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}
J. Kuruvilla, P. Armand, A. F. Herrera, V. Ribrag, C. Thieblemont, B. von Tresckow, S. Thompson, K. E. Ryland, R. Z. Yusuf, P. L. Zinzani
{"title":"FINAL SAFETY ANALYSIS IN PARTICIPANTS WITH HEMATOLOGIC MALIGNANCIES WHO RECEIVED ALLOGENEIC STEM CELL TRANSPLANT AFTER PEMBROLIZUMAB THERAPY","authors":"J. Kuruvilla, P. Armand, A. F. Herrera, V. Ribrag, C. Thieblemont, B. von Tresckow, S. Thompson, K. E. Ryland, R. Z. Yusuf, P. L. Zinzani","doi":"10.1002/hon.70094_351","DOIUrl":"https://doi.org/10.1002/hon.70094_351","url":null,"abstract":"<p><b>Introduction:</b> Allogeneic stem cell transplant (allo-SCT) carries a relevant risk of transplantation-related mortality (TRM), especially from graft-versus-host disease (GVHD), particularly in patients previously treated with checkpoint inhibitors. Final safety results in participants (pts) with hematologic malignancies who received allo-SCT after pembrolizumab (pembro) therapy across a variety of pembro clinical studies are presented.</p><p><b>Methods:</b> Data were pooled from 10 phase 1–3 studies (KEYNOTE-A33 [NCT04317066], KEYNOTE-013 [NCT01953692], KEYNOTE-155 [NCT02684617], KEYNOTE-051 [NCT02332668], KEYNOTE-B68 [NCT04875195], KEYNOTE-087 [NCT02453594], KEYNOTE-170 [NCT02576990], KEYNOTE-183 [NCT02576977], KEYNOTE-204 [NCT02684292], and MK4280-003 [NCT03598608]). Outcomes of interest included acute and chronic GVHD, incidence of allo-SCT–related adverse events, overall survival (OS), and TRM.</p><p><b>Results:</b> A total of 112 pts were reported to have received allo-SCT, 14 were not included in the analysis (13 received allo-SCT > 2 years after last dose of pembro and 1 had a missing date of allo-SCT). Median duration on study treatment was 5.6 months (range, 0.03–29.7), and median time from last dose of pembro to first allo-SCT was 4.9 months (range, 1–20). Of 98 evaluable pts, 67 pts (68%) received intervening therapy between pembro and allo-SCT. At time of transplant, 47 pts (48%) had active disease, 39 pts (40%) were in remission, and 12 (12%) had unknown disease status. A total of 91 pts (93%) received allo-SCT only; 7 (7%) received autologous SCT followed by allo-SCT. Among 63 pts (64%) who developed GVHD, 52 (53%) experienced acute events (Glucksberg II-IV [<i>n</i> = 34] and Glucksberg III-IV [<i>n</i> = 19]) and 24 (25%) experienced chronic events (mild [<i>n</i> = 11], moderate [<i>n</i> = 8], and severe [<i>n</i> = 5]). The most common sites for chronic GVHD were skin (<i>n</i> = 15), oral mucosa (<i>n</i> = 12), and liver (<i>n</i> = 9). Other predetermined non-GVHD events of clinical interest occurred in 42 pts (43%), categorized as critical illness (30%), febrile syndrome treated with corticosteroids (1%), immune-mediated adverse events (10%), pulmonary complications (14%), and venoocclusive liver disease (3%). The most common predetermined events of clinical interest (≥ 5%) were febrile neutropenia and pneumonia (6% each). The most common of the other adverse events not included in the 5 categories above (≥ 2%) were grade ≤ 2 pyrexia (5%) and rash (2%). Median OS was not reached (NR; 95% CI: NR-NR) post–allo-SCT, with 100-day, 24-month, and 48-month OS rates of 94%, 70%, and 68%, respectively. Estimated TRM rates at 100 days, 24 months, and 48 months were 5%, 21%, and 21%, respectively.</p><p><b>Conclusion:</b> The inclusion of additional pts in this analysis revealed comparable rates of acute and chronic GVHD, OS, and TRM compared to previous studies and historical benchmarks, thus, reinforcing the role of allo-","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_351","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292664","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}
E. Corvilain, R. Di blasi, C. Thieblemont, M. Cheminant, B. Guffroy, J. Decroocq, A. Campidelli, M. Rubio, F. Claves, S. Carras, M. Mothy, V. Dupont, R. Houot, S. Choquet
{"title":"CD19 CAR-T CELL THERAPY IN RELAPSED/REFRACTORY SOLID ORGAN TRANSPLANT-RELATED LYMPHOPROLIFERATION: A LYSA ANALYSIS OF THE FRENCH COHORT DESCAR-T","authors":"E. Corvilain, R. Di blasi, C. Thieblemont, M. Cheminant, B. Guffroy, J. Decroocq, A. Campidelli, M. Rubio, F. Claves, S. Carras, M. Mothy, V. Dupont, R. Houot, S. Choquet","doi":"10.1002/hon.70094_381","DOIUrl":"https://doi.org/10.1002/hon.70094_381","url":null,"abstract":"<p><b>Introduction:</b> Post-transplant lymphoproliferative disorder (PTLD) is a rare but severe complication of solid organ transplantation (SOT). First-line treatment typically involves reducing immunosuppressive drugs (ID) alongside Rituximab, followed by maintenance for partial responders (PR) or immunochemotherapy for progression disease (PD). The management of refractory/relapsed (R/R) remains uncertain. The role of CD19 CAR-T cells is not established, though case reports and a series of 22 patients have been published.</p><p><b>Methods:</b> We conducted a retrospective analysis of the multicenter French DESCAR-T registry (NCT04328298) to identify patients treated by CD19 CAR-T cells for SOT-related PTLD. The study period spanned from July 2019 to September 2024. Survival analyses were performed using Kaplan-Meier models.</p><p><b>Results:</b> We identified 12 patients (5 males) treated by CD19 CAR-T cells. Prior SOTs included kidneys (<i>n =</i> 10), liver (<i>n =</i> 1) and lungs (<i>n =</i> 1). In all but one patient, lymphoproliferation occurred more than one year after SOT. All PTLD were classified as diffuse large B cell lymphoma (EBV-associated in 10/11, 1 not available (NA)), except one diagnosed as transformed marginal zone lymphoma. CAR-T cells were administered as second line treatment in 3 patients, and beyond second line in 9 patients. The median age at infusion was 41 years (IQR: 22–62). At diagnosis, performance status was 0-1 in 11/12, Ann-Arbor stage was III-IV in 8/12 and aaIPI score was 1-2 in 11/12 patients. The median lymphocyte rate at apheresis was 0.6G/L (IQR:0-3.4). CAR-T products used included axicabtagene ciloleucel (<i>n =</i> 9) and tisagenlecleucel (<i>n =</i> 3). Ten patients received cyclophosphamide-fludarabine as conditioning regimen (2 NA). ID was modified in all patients at diagnosis then before apheresis. At CAR-T infusion, patients were receiving corticosteroids alone (<i>n</i> = 8), m-TOR inhibitor (<i>n</i> = 1), corticosteroids and calcineurin inhibitor (<i>n =</i> 1), or no ID (<i>n =</i> 2). Cytokine-release-syndrome occurred in all patients with grade 3–4 in 2 cases. Immune-effector-cell associated neurotoxicity (ICANS) was observed in 7 patients with grade 3–4 in 2 cases. Hypogammaglobulinemia (< 5 g/L) was reported in 70% of patients (7/10) without need of immunoglobulins replacement. The best ORR was 82%, including 64% complete response, 18% PR and 18% PD. With a median follow-up of 12.9 months (mo) (95% CI: 4.5-(-)), median progression-free-survival and overall survival were both 16.6 mo (95% CI: 1.1-(-) and 2-(-), respectively) (Figure). Six patients died among which three due to PD and 2 from high grade ICANS (one acute, one late) (1 NA). Among the six surviving patients, one experienced kidney rejection requiring a return to dialysis.</p><p><b>Conclusion:</b> This is the second reported series of patients with R/R SOT-related PTLD treated with CD19 CAR-T cells. Our findings suggest that ","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_381","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292685","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}
N. Dakhallah, J. Steglich, A. L. Alazraki, S. M. Castellino, K. Dieckmann, J. E. Flerlage, C. Gowdy, M. B. Heneghan, K. M. Kelly, H. A. Lai, C. Mauz-Körholz, K. M. McCarten, S. Milgrom, R. Pabari, M. Palese, S. D. Voss, L. Kurch, D. Stoevesandt, J. Seelisch
{"title":"IMAGING PITFALLS IN PEDIATRIC, ADOLESCENT, AND YOUNG ADULT HODGKIN LYMPHOMA: A SEARCH FOR CAYAHL INITIATIVE TO BRIDGE MULTIDISCIPLINARY PATIENT CARE","authors":"N. Dakhallah, J. Steglich, A. L. Alazraki, S. M. Castellino, K. Dieckmann, J. E. Flerlage, C. Gowdy, M. B. Heneghan, K. M. Kelly, H. A. Lai, C. Mauz-Körholz, K. M. McCarten, S. Milgrom, R. Pabari, M. Palese, S. D. Voss, L. Kurch, D. Stoevesandt, J. Seelisch","doi":"10.1002/hon.70094_374","DOIUrl":"https://doi.org/10.1002/hon.70094_374","url":null,"abstract":"<p>N. Dakhallah and J. Steglich equally contributing author.</p><p><b>Introduction:</b> Hodgkin lymphoma (HL) is a highly curable malignancy in children, adolescents, and young adults (CAYA), and current treatment strategies aim to minimize adverse late effects. Many patients are enrolled in clinical trials that include centralized review for both initial and interim staging. While academic guidelines provide a structured framework, real-world clinical scenarios sometimes present imaging pitfalls that require nuanced judgment.</p><p><b>Methods</b>: The Staging, Evaluation and Response Criteria (SEARCH) for CAYAHL initiative was established in 2011 to harmonize staging and response criteria in HL in CAYA. However, applying these published criteria can present challenges in situations where imaging pitfalls are encountered. This SEARCH for CAYAHL project is a transatlantic collaboration among experts in diagnostic radiology, nuclear medicine radiology, radiation oncology and pediatric oncology. A working group first identified the most frequent and relevant imaging pitfalls in HL. Through literature review, imaging and clinical experience, and the use of specific real-word cases, this effort defines and describes imaging pitfalls in HL in CAYA.</p><p><b>Results</b>: Morphologic and metabolic imaging pitfalls in HL refer to the misinterpretation of findings that can occur during staging, disease evaluation, or post-treatment surveillance. These radiological findings are not indicative of disease but are rather manifestations of other causes that are specific to each tissue or organ. In this work, we explore pitfalls resulting from suboptimal imaging conditions, concurrent inflammatory, infectious, or other causes. We discuss organ specific pitfalls involving the lymph nodes, lungs, bone, bone marrow, spleen, liver and Waldeyer’s ring and present important considerations on imaging following the completion of therapy.</p><p><b>Conclusions</b>: This collaborative effort aims to disseminate insights gained from decades of centralized review experience in North American and European trials to optimize patient care by integrating imaging and clinical expertise. Although not intended as a comprehensive staging guide, it highlights recurrent imaging pitfalls that may lead to diagnostic uncertainty. By encouraging interdisciplinary exchange, this work seeks to complement existing literature and serve as a troubleshooting guide for situations where clinical realities diverge from academic paradigms, ultimately paving the way for improved patient outcomes.</p><p><b>Keywords:</b> Hodgkin lymphoma (pediatric, adolescent, and young adult)</p><p><b>Potential sources of conflict of interest:</b></p><p><b>C. Mauz-Körholz</b></p><p><b>Employment or leadership position:</b> Institutional research grant Merck</p><p><b>Consultant or advisory role:</b> Merck advisory board</p>","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_374","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292688","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}
S. K. Thiruvengadam, L. Chen, S. Buller, V. Iyengar, G. Pelaez, V. Pizzuti, A. Major, Y. Youssef, Y. Sawalha, D. Wallace, M. Masterson, N. Birrer, A. Bock, A. Nedved, Y. Wang, T. Lucido, J. Rhodes, J. Crombie, D. Montana, M. Stanchina, J. P. Alderuccio, A. Gibson, P. A. Riedell, T. Jain, B. Heyman, H. Rasmussen, C. Ujjani, P. Gould, H. Cherng, A. Bahnasy, T. Hilal, B. Parker, M. A. Moustafa, S. Pak, M. Okwali, J. Chicola, J. M. Manzano, C. Goth, D. Russler-Germain, P. Torka, A. F. Herrera
{"title":"MULTICENTER REAL-WORLD OUTCOMES OF FRONTLINE POLA-R-CHP IN TREATMENT NAÏVE DLBCL","authors":"S. K. Thiruvengadam, L. Chen, S. Buller, V. Iyengar, G. Pelaez, V. Pizzuti, A. Major, Y. Youssef, Y. Sawalha, D. Wallace, M. Masterson, N. Birrer, A. Bock, A. Nedved, Y. Wang, T. Lucido, J. Rhodes, J. Crombie, D. Montana, M. Stanchina, J. P. Alderuccio, A. Gibson, P. A. Riedell, T. Jain, B. Heyman, H. Rasmussen, C. Ujjani, P. Gould, H. Cherng, A. Bahnasy, T. Hilal, B. Parker, M. A. Moustafa, S. Pak, M. Okwali, J. Chicola, J. M. Manzano, C. Goth, D. Russler-Germain, P. Torka, A. F. Herrera","doi":"10.1002/hon.70094_289","DOIUrl":"https://doi.org/10.1002/hon.70094_289","url":null,"abstract":"<p><b>Introduction:</b> Incorporation of polatuzumab vedotin into initial therapy of diffuse large B-cell lymphoma (DLBCL) became a standard of care (SOC) based on the POLARIX trial, which demonstrated improvement in progression free survival (PFS) with pola-R-CHP compared to R-CHOP. In this study we evaluate the safety and efficacy of SOC frontline pola-R-CHP for treatment naïve DLBCL.</p><p><b>Methods:</b> We performed a multicenter retrospective study including patients (pts) from 17 US centers. Pts with treatment naïve DLBCL who received pola-R-CHP as frontline therapy outside of the setting of a clinical trial were eligible. The primary objective was to evaluate the PFS of SOC pola-R-CHP in frontline DLBCL. Secondary objectives included evaluating the safety of the regimen as well as secondary measures of efficacy such as overall response rate (ORR), complete response rate (CRR), overall survival (OS), and time to next treatment (TTNT).</p><p><b>Results:</b> A total of 535 pts treated with pola-R-CHP between August 2021 to September 2024 were included. At least one cycle of alternate treatment was given in 28% pts before switching to pola-R-CHP (78% of these pts received R-CHOP). The median age was 67 years (range 22–90), 41% were female, 17% had ECOG ≥ 2, 89% had advanced stage, 45% had > 1 extranodal site, 63% had elevated LDH, 63% had an IPI of 3–5, 33% had bulky disease (≥ 7.5 cm), 3.3% had central nervous system involvement, 29% were double expressor, 3.9% were double/triple hit, 34% had germinal center B-cell subtype (GCB) and 59% had non-GCB subtype by Hans.</p><p>ORR was 92% with a CRR of 80% in all pts; ORR was 93% versus 92% (CRR 77% vs. 81%) for GCB versus non-GCB. At a median follow up of 11 months (range 0.5–32), 1-year PFS was 81% (95% CI: 77%–84%) among all pts, 78% for GCB versus 82% for non-GCB; 1-year OS was 91% (95% CI: 87%–93%) among all pts and 91% for both cell of origin (COO) subgroups. Subsequent treatment was given in 16% pts with median TTNT 5.8 months (range 0.8–18).</p><p>With respect to safety, 37% developed any grade neuropathy with 1.1% grade ≥ 3, 20% had grade ≥ 3 infection, 3.4% had cardiomyopathy, 31% had grade ≥ 3 neutropenia, 15% had grade ≥ 3 febrile neutropenia, and 15% had grade ≥ 3 thrombocytopenia. 31% were hospitalized and 6.0% had ICU admission for treatment-related adverse events. Treatment was discontinued in 13% due to toxicity (4.5%), progression (3.6%), or other reasons (4.7%). Seven deaths (1.3%) were deemed to be related to pola-R-CHP.</p><p>In univariate Cox models, > 1 extranodal sites, bulky disease, CNS involvement, and double/triple hit were associated with inferior PFS (<i>p</i> < 0.05) and ECOG ≥ 2, elevated LDH, and IPI 3–5 were associated with inferior OS and PFS (<i>p</i> < 0.05). COO was not significantly associated with PFS or OS.</p><p><b>Conclusion:</b> The results of our study suggest that SOC pola-R-CHP is safe and effective for treatment naïve DLBCL, with outcomes","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_289","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292724","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}
D. Baggio, S. Han, P. Ghione, I. Okcu, J. Okosun, M. Kamdar, A. Kuhnl, N. Grover, H. Cherng, A. Olszewski, P. Ramakrishnan Geethakumari, J. Smith, A. Prica, J. Khwaja, W. Osborne, J. Koff, D. El-Sharkawi, N. Wagner-Johnston, J. Munoz, A. Desai, N. Epperla, F. Baidoun, T. Moyo, M. Narkhede, A. Barrett, K. Linton, D. Wallace, N. Elmusharaf, J. Sandoval-Sus, A. Danilov, J. Rhodes, A. Santarsieri, F. Karim, J. Calabrese De Feo, R. Tavarozzi, I. Nizamuddin, N. Dong, A. Saha, W. Hann, Y. Wang, R. Treitman, A. Maraj, S. Tolu, T. Ollila, C. Abeyakoon, V. Calvert, A. Ayers, A. Hilali, I. Trutzer, S. Monick, J. Sharp, H. Tun, N. Ghosh, G. P. Collins, V. Deshani, J. Can, A. Kallam, B. Kahl, J. Chavez, K. Cwynarski, J. P. Alderuccio
{"title":"OUTCOMES IN AN INTERNATIONAL MULTICENTRE STUDY OF 600 PATIENTS WITH CNS RELAPSE OF LARGE B CELL LYMPHOMA: COMPARTMENT OF RELAPSE IS PROGNOSTIC","authors":"D. Baggio, S. Han, P. Ghione, I. Okcu, J. Okosun, M. Kamdar, A. Kuhnl, N. Grover, H. Cherng, A. Olszewski, P. Ramakrishnan Geethakumari, J. Smith, A. Prica, J. Khwaja, W. Osborne, J. Koff, D. El-Sharkawi, N. Wagner-Johnston, J. Munoz, A. Desai, N. Epperla, F. Baidoun, T. Moyo, M. Narkhede, A. Barrett, K. Linton, D. Wallace, N. Elmusharaf, J. Sandoval-Sus, A. Danilov, J. Rhodes, A. Santarsieri, F. Karim, J. Calabrese De Feo, R. Tavarozzi, I. Nizamuddin, N. Dong, A. Saha, W. Hann, Y. Wang, R. Treitman, A. Maraj, S. Tolu, T. Ollila, C. Abeyakoon, V. Calvert, A. Ayers, A. Hilali, I. Trutzer, S. Monick, J. Sharp, H. Tun, N. Ghosh, G. P. Collins, V. Deshani, J. Can, A. Kallam, B. Kahl, J. Chavez, K. Cwynarski, J. P. Alderuccio","doi":"10.1002/hon.70094_339","DOIUrl":"https://doi.org/10.1002/hon.70094_339","url":null,"abstract":"<p>D. Baggio, S. Han, K. Cwynarski, and J. P. Alderuccio equally contributing author.</p><p><b>Background</b>: Secondary central nervous system lymphoma (SCNSL) is a rare disease defined by involvement of the CNS concurrent to or following a diagnosis of systemic large B-cell lymphoma (LBCL). This international multicentre cohort study aimed to determine survival outcomes in patients (pts) with isolated CNS relapse (RI-SCNSL) and synchronous systemic/CNS relapse (RS-SCNSL).</p><p><b>Methods:</b> Pts ≥ 18 years treated 2001–2023 at 35 US, UK and Canadian centres were included. CNS involvement was categorised as: brain parenchyma, spine, leptomeninges, vitreoretinal, or > 1 compartment. The primary outcomes were time-to-CNS event from LBCL diagnosis, and progression-free and overall survival (PFS and OS) from time of CNS involvement.</p><p><b>Results:</b> Six hundred pts were included: 393 RI-SCNSL and 207 RS-SCNSL. Prior systemic LBCL treatment was RCHOP (450; 75%) or REPOCH (82; 14%) in the majority; median age at LBCL diagnosis was 62 (range 53–69); and CNS IPI was 4–6 in 139 (23%). Intrathecal and systemic methotrexate prophylaxis was delivered in 40 (10%) and 44 (11%) RI-SCNSL, and 29 (14%) and 33 (16%) RS-SCNSL respectively. Of RI-SCNSL, involvement of parenchyma (<i>n</i> = 257; 65%), leptomeninges (62; 16%), spine (14; 3.5%), vitreoretinal (11; 2.8%) and > 1 compartment (49; 2.5%) were observed. Of RS-SCNSL, these represented 77 (37%), 77 (37%), 15 (7.2%), 2 (1.0%) and 36 (17%), respectively.</p><p>Median time-to-CNS event was 10.3 months (range 6.6–25.6) for RI-SCNSL and 8 months (5.5–16.5) for RS-SCNSL (<i>p</i> = 0.0001). Median PFS and OS were superior for RI-SCNSL (14.7 and 22.6 months) compared to RS-SCNSL (6.8 and 8.2 months, <i>p</i> < 0.0001). Median time-to-CNS event was longer in both RI-SCNSL (<i>p</i> < 0.0001) and RS-SCNSL (<i>p</i> = 0.019) for parenchymal (13.1 and 10.9 months for RI and RS-SCNSL) and vitreoretinal disease (10.8 and 27.1 months for RI and RS-SCNSL) than leptomeningeal disease (6.9 months in both groups). In RI-SCNSL, CNS compartment was prognostic of PFS (<i>p</i> = 0.023), longest for vitreoretinal (median 117.4 months) and shortest for leptomeningeal disease (median 7.6 months). Outcomes were poor irrespective of CNS compartment in RS-SCNSL.</p><p>Comparing 139 patients with leptomeningeal to 257 with parenchymal involvement, leptomeningeal disease was enriched for <i>MYC/BCL2</i> double-hit status (20% versus 6.3% of parenchymal, <i>p</i> = 0.0002), ≥ 3 extranodal sites at LBCL diagnosis (17% versus 9.3%, <i>p</i> = 0.013), and bone marrow involvement (37% versus 17%, <i>p</i> < 0.0001). Conversely, isolated parenchymal involvement was enriched for a history of testicular LBCL (12% versus 3.6%, <i>p</i> = 0.017).</p><p><b>Conclusion:</b> Compartment of relapse holds prognostic significance in SCNSL. PFS and OS are significantly better for RI-SCNSL compared to RS-SCNSL. Isolated vitreoretina","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_339","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292824","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}
E. Lucchini, R. Sciarra, V. Tomarchio, S. Pelliccia, M. Marangon, E. Maiolo, M. Novo, L. Schiattone, M. Farina, G. Scapinello, F. M. Quaglia, J. Olivieri, L. Pagliaro, P. Angelillo, L. Mannelli, C. Ghiggi, F. Zaja
{"title":"THE ROLE OF STEM CELL TRANSPLANTATION IN PATIENTS WITH R/R CLASSICAL HODGKIN LYMPHOMA TREATED WITH CHECKPOINT INHIBITORS: ITALIAN MULTICENTER EXPERIENCE","authors":"E. Lucchini, R. Sciarra, V. Tomarchio, S. Pelliccia, M. Marangon, E. Maiolo, M. Novo, L. Schiattone, M. Farina, G. Scapinello, F. M. Quaglia, J. Olivieri, L. Pagliaro, P. Angelillo, L. Mannelli, C. Ghiggi, F. Zaja","doi":"10.1002/hon.70094_364","DOIUrl":"https://doi.org/10.1002/hon.70094_364","url":null,"abstract":"<p>There is no agreement on the role and positioning of stem cell transplantation (SCT) in patients (pts) with relapsed/refractory (R/R) classical Hodgkin lymphoma (cHL) who undertake treatment with checkpoint inhibitors (CPI). The aim of this study is to evaluate the real-world outcomes of R/R cHL pts treated with CPI, particularly the use of consolidation with autologous (autoSCT) and allogeneic SCT (alloSCT). Observational, retrospective, multicenter study which enrolled consecutive pts with R/R cHL aged 18–70 years, treated with CPI monotherapy in Italy from January 2016 to June 2023. Primary objective was the proportion of pts who received consolidation with alloSCT.</p><p>We enrolled 126 pts from 15 Italian centers. Median age at CPI start was 36 years (18–70), 54% males, 95% received first-line ABVD, 51% previously received autoSCT and 98% brentuximab vedotin. Eighty-six pts (68%) were treated with pembrolizumab, 40 (32%) with nivolumab. Overall response rate to CPI was 81%, with 49% complete responses (CRs). 107 pts (85%) were considered fit for alloSCT, of those 62 (58%) were planned to receive alloSCT. Ultimately, 41 pts (32%) received alloSCT, 36 (29%) autoSCT and 49 (39%) received no consolidation. Reasons for not performing alloSCT in candidates were progressive disease (PD) (<i>n</i> = 9), donor unavailability (<i>n</i> = 4), patient’s refusal or comorbidities (<i>n</i> = 4), others (<i>n</i> = 4). Twenty-nine out of 41 allo-transplanted pts (70%) had already received an autoSCT before CPI. Reasons for not administering a consolidation after CPI were age/comorbidity (<i>n</i> = 19), center’s choice (<i>n</i> = 9), alloSCT refusal or donor unavailability (<i>n</i> = 9), PD (<i>n</i> = 7), other (<i>n</i> = 5).</p><p>In pts who consolidated with alloSCT or autoSCT, median PFS and OS were not reached at a median follow-up of 27 (range 7–90) and 47.5 months (range 4–99) from CPI treatment, respectively. 48-months PFS probability were 80.6% and 81% for alloSCT and autoSCT recipients. Transplant-related mortality after alloSCT was 14.6%. In non-consolidated pts, at a median follow-up of 21 months, median PFS was 27 months. Seventeen pts (35%) were still receiving CPI at last follow-up; the main reason for interruption was PD, in 18/32 pts (56%). In pts who received SCT consolidation there was no difference in terms of PFS according to the obtainment of a CR with CPI (<i>p</i> = 0.93 for alloSCT and <i>p</i> = 0.6 for autoSCT). Instead, pts who didn’t consolidate had a significantly poorer outcome if they didn’t obtain a CR with CPI (median PFS not reached versus 11 months; <i>p</i> < 0.001)—Figure 1.</p><p>Outcomes after autoSCT and alloSCT consolidation are excellent, regardless of the response achieved with CPI; alloSCT was burdened by higher toxicity. Pts who achieved a CR with CPI showed good outcomes even without consolidation, however autoSCT should be considered if not performed before. Pts who do not achieve/loss the CR and hav","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_364","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292825","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}
W. S. Kim, S. J. Kim, D. H. Yoon, H. W. Cho, S. E. Yoon, D. H. Yang, H. Shin, H. S. Eom, E. Y. Lee, J. M. Byun, Y. I. Koh, D. Y. Shin, J. Hong, H. W. Lee, J. H. Jung, S. S. Yoon, G. Y. Song, D. Y. Kim
{"title":"ANBALCABTAGENE AUTOLEUCEL (PD-1 AND TIGIT KNOCKDOWN CD19 CAR-T) FOR RELAPSED/REFRACTORY LARGE B-CELL LYMPHOMA (CRC01-01)","authors":"W. S. Kim, S. J. Kim, D. H. Yoon, H. W. Cho, S. E. Yoon, D. H. Yang, H. Shin, H. S. Eom, E. Y. Lee, J. M. Byun, Y. I. Koh, D. Y. Shin, J. Hong, H. W. Lee, J. H. Jung, S. S. Yoon, G. Y. Song, D. Y. Kim","doi":"10.1002/hon.70094_319","DOIUrl":"https://doi.org/10.1002/hon.70094_319","url":null,"abstract":"<p><b>Introduction:</b> Anbal-cel is CD19 CAR-T with PD-1, TIGIT knockdown to reduce the negative impact on response. This approach is expected to exert a stronger and more sustained effect on tumor cells.</p><p><b>Methods:</b> In this multicenter, phase 2 trial, we enrolled adult patients with relapsed or refractory large B-cell lymphomas including DLBCL-NOS, HGBL, PMBCL, FL3B. Lymphodepletion was performed with cyclophosphamide 500 mg/m<sup>2</sup> and fludarabine 30 mg/m<sup>2</sup>, administered for three days (-5, -4, -3 days). The median dose was 2.2 ×10<sup>6</sup>cells/kg (range, 0.2–2.61 (×10<sup>6</sup>cells/kg)). The primary endpoint is ORR by independent review committee. The secondary endpoints are PFS, OS and safety.</p><p><b>Results:</b> Between January 2022 and September 2023, 91 patients underwent leukapheresis to manufacture and 79 patients received anbal-cel. Most patients were DLBCL, NOS (94.9%) and 65 years or older accounted for 53.2%. The patients who had received up to second-line chemotherapy was 70.9%. The IPI score was distributed as follows: low in 31.6%, low-intermediate in 25.3%, high-intermediate in 34.2%, high in 8.9%. The median SPD was 1858 mm<sup>2</sup> (range, 150–13,724), and the median TMTV was 63.3 ml (range, 0–3603). 26.3% of patients had GCB phenotype, and 61.8% were classified as double expressors.</p><p>73 patients included in an efficacy analysis set. The ORR was 75.3% and CR rate was 67.1%. The median f/u time was 8.5 months (range, 0.2–22.6). The median PFS was 6.0 months (95% CI: 4.3–16.5 months), with 12-month and 18-month PFS rates of 41.1% and 35.2%, respectively. The median OS was not reached, and the 12-month and 18-month OS rates were 66.6% and 57.3%, respectively. Regarding CAR-T cell expansion, the responder group exhibited a significantly higher peak expansion (median C<sub>max</sub>: 22,281 copies/µg) compared to the non-responder group (median C<sub>max</sub>: 9781 copies/µg).</p><p>We categorized patients into long-term responders (LR) and non-long-term responders (non-LR) based on whether they maintained CR at 6 months. FACS analysis showed that PD-1 and TIGIT expression on anbal-cel was significantly lower in the LR group compared to the non-LR group, with this difference persisting at days 14. Additionally, CAR-T cells in the LR group displayed a less activated memory phenotype, characterized by lower expression of CD39 and Tim-3 and higher expression of CD127, compared to those in the non-LR group. Consistent with this, the non-LR group exhibited an increased frequency of terminally differentiated and functionally senescent CAR-T cells, marked by the expression of CD57, CD38, and HLA-DR.</p><p>CRS and neurologic events occurred in 57.0% and 13.9%, respectively. Grade 3 CRS occurred in 8.9% with no cases of grade 4. Neurologic events of grade 3 or higher were reported in 3.8%. The rate of serious infections was 25%, prolonged cytopenia was reported in 44% of patients.</p><p><b>Conclus","PeriodicalId":12882,"journal":{"name":"Hematological Oncology","volume":"43 S3","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/hon.70094_319","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144292964","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}