September 7, 2025
Lymphoma News SOHO 2025 News

Megan Melody on HL and MCL research at SOHO 2025

Megan Melody, MD, is an assistant professor at the University of South Florida and lymphoma and CAR T-cell therapy specialist at Tampa General Hospital Cancer Institute. Here’s what she’s looking forward to when it comes to Hodgkin lymphoma and mantle cell lymphoma research at the meeting.

Hodgkin lymphoma

There are so many exciting and impactful Hodgkin lymphoma (HL) posters to check out at this year’s poster sessions. With a focus on cutting-edge research, the session will feature a series of abstracts that address critical challenges in managing this complex disease.

Among the posters to be presented, three stand out among the highest-rated abstracts submitted for presentation at SOHO this year for their potential to reshape clinical practice and deepen our understanding of HL, spanning the use of circulating tumor DNA (ctDNA) as a biomarker, the cardiovascular risks of immune checkpoint inhibitors, and the management of treatment-related cardiotoxicity.

One of the most promising developments in the treatment landscape for classical HL (cHL) is the utilization of ctDNA as a biomarker to assess disease burden and monitor treatment response. As the application of checkpoint inhibitors such as pembrolizumab and nivolumab continues to expand for the management of cHL, ctDNA analysis offers additional practical advantages, especially given the complexities and sometimes ambiguous interpretations associated with PET/CT scans in this setting.

A study from Ryan Lynch, MD, focuses on early-stage, non-bulky cHL treated with a combination regimen of brentuximab vedotin, nivolumab, doxorubicin, and dacarbazine. Using an ultrasensitive PhasEDseq ctDNA measurable residual disease assay, the researchers evaluated the correlation between dynamic changes in ctDNA levels and treatment outcomes evaluated by PET/CT scan.

The study demonstrated that nearly all patients (94%) exhibited detectable baseline ctDNA levels, which were strongly correlated with the overall disease burden as assessed by stage and International Prognostic Index score. Notably, treatment resulted in a rapid reduction in ctDNA, with levels becoming undetectable by the conclusion of therapy. This early decline in ctDNA levels often preceded the achievement of complete metabolic responses as evidenced by PET/CT imaging, suggesting that ctDNA clearance may serve as an early and highly sensitive indicator of treatment efficacy.

Such findings support the integration of ctDNA as an adjunct to traditional imaging modalities, potentially enhancing the precision of response assessments in clinical practice. Furthermore, the ability of ctDNA to provide real-time insights into tumor dynamics highlights its potential to transform patient monitoring and guide more tailored treatment strategies. Dr. Lynch’s presentation is sure to spark discussion on how to integrate this emerging biomarker into routine clinical practice.

Cardiotoxicity is a recognized complication in the treatment of lymphoma, especially among patients receiving anthracycline-based chemotherapy. Despite this, there is limited data on the true incidence of cardiotoxicity and the identification of patients at highest risk for developing these adverse effects.

At this year’s poster session, a single-center retrospective analysis involving 349 lymphoma patients with a minimum of five years of post-treatment follow-up aims to quantify both the incidence and the risk factors for chemotherapy-induced cardiotoxicity, with particular emphasis on patients with HL.

The study found that the overall incidence of cardiotoxicity was 1.72%, with advanced age (≥70 years) being a significant risk factor. While other factors such as diabetes, hypertension, and treatment intensity showed higher rates of cardiotoxicity, these were not statistically significant. This study highlights the importance of vigilant cardiac monitoring, particularly in older patients and those with preexisting cardiovascular risk factors and is a timely reminder of the need for comprehensive risk assessment and proactive management of cardiotoxicity in lymphoma patients.

Although the cardiac toxicities associated with anthracycline-based regimens are well documented, the cardiovascular risks linked to immune checkpoint inhibitors (ICIs), particularly PD-1 inhibitors such as pembrolizumab and nivolumab, are less clearly defined.

Among the posters presentations this year, one study presents a comprehensive analysis of the FDA Adverse Event Reporting System database, specifically examining the risk of cardiovascular events such as myocardial infarction and stroke in patients treated with ICIs.

The analysis revealed that PD-1 inhibitors (pembrolizumab and nivolumab) were associated with a significantly increased risk of stroke while other ICIs (such as atezolizumab, avelumab, durvalumab, ipilimumab, and tremelimumab) showed no such association with increased incidence of cardiovascular or atherosclerotic events. It is important to note that these other agents are used infrequently in the treatment of cHL, and the absolute number of patients treated with these medications was not reported, which may impact the interpretation of these findings.

Regardless, this study underscores the consideration that as the treatment paradigm for lymphoma shifts from traditional chemotherapy-based regimens to immunotherapy of ICI-based treatment strategies, clinicians may be trading one class of treatment-related toxicities for another. Consequently, as ICIs become more widely adopted, understanding and mitigating their cardiovascular risks will be essential to optimizing patient outcomes. The presentation will likely prompt discussion on how to integrate cardiovascular risk assessments into the management of patients receiving ICIs.

These abstracts highlight just a few examples of the impactful research being presented at this year’s SOHO meeting poster session, which promises to be a dynamic and thought-provoking exploration of the latest advancements in diagnosis, treatment, and patient care in the field of cHL.

Mantle cell lymphoma

This year’s poster session is sure to include some cutting-edge research in the field of mantle cell lymphoma (MCL) and promises to shed light on some of the latest developments in treatment strategies.

The session will feature research examining the incorporation of novel therapies, in particular bispecific antibodies (BsABs) into the treatment paradigm for relapsed/refractory (R/R) MCL, as well as advancements in frontline management for this highly treatable disease.

In the frontline setting, Martin Dreyling, MD, will present a subset analysis of the phase 3 ECHO trial (NCT02972840), which demonstrated that acalabrutinib combined with bendamustine-rituximab (ABR) significantly improved progression-free survival (PFS) compared with bendamustine-rituximab (BR) alone in older patients (≥65 years) with decreased performance status and previously untreated MCL.

Dr. Dreyling and colleagues examined a subset of patients with high-risk MCL, defined as a lymphoma IPI score of 6-11, presence of TP53 mutations, Ki-67 index ≥30%, and/or blastoid or pleomorphic histology. Notably, over 60% of patients in both treatment arms were identified as having high-risk disease, emphasizing the high incidence of poor-risk MCL in the older patient population and the importance of this analysis.

The results showed that the overall response rate (ORR), complete response (CR), and median PFS were all significantly better with ABR compared with BR in this high-risk population. Though overall survival (OS) appeared similar between the groups, the marked improvement in PFS underscores the potential of ABR as a more effective frontline treatment option for high-risk MCL.

These findings are particularly relevant as the oncology community continues to explore strategies to improve outcomes in this particularly vulnerable and treatment-resistant population and is sure to spark discussion on how the data should impact clinical practice.

Included in this year’s most impactful abstracts are two studies that further exemplify the paradigm shift in the treatment landscape of R/R MCL toward the integration of novel CD20xCD3 BsAbs. Both glofitamab and mosunetuzumab are BsAbs, and their remarkable efficacy in R/R MCL underscores the transformative potential of this therapeutic class.

In the first abstract, Tycel Phillips, MD, expands upon his previously published findings from a phase 1/2 clinical trial investigating glofitamab monotherapy in heavily pretreated R/R MCL patients. The study particularly emphasizes the efficacy of glofitamab in patients with high-risk disease based on histopathological features. Nearly half (45.6%) of the evaluable patients had at least one high-risk histological feature, such as Ki-67 proliferation index ≥50%, blastoid or pleomorphic variants, or TP53 mutations.

Remarkably, 81% of patients with high-risk features achieved a CR compared with 68% in patients without such features. These findings highlight the potency of glofitamab in a heavily pretreated cohort with aggressive disease biology.

Furthermore, Michael Wang, MD, presented a pre-planned cohort analysis of a phase 2 trial evaluating a novel combination therapy of mosunetuzumab plus the antibody-drug conjugate polatuzumab vedotin (Mosun+Pola). This combination has already demonstrated remarkable efficacy with minimal toxicity for the treatment of diffuse large B-cell lymphoma (Budde et al. Nat Med. 2023.).

The data presented at SOHO 2025 further highlights the efficacy of this treatment in MCL. In this study, 42 patients with high-risk features (including advanced stage, elevated MCL IPI score, Ki-67 ≥50%, blastoid or pleomorphic variants, or TP53 mutations) were treated with Mosun+ Pola. Patients were heavily pretreated, having received a median of three prior lines of therapy; 26% previously received chimeric antigen receptor T-cell therapy, and 93% were refractory to their last prior line of therapy.

The results revealed an ORR of 88% and a CR rate of 79%, with a median PFS of 19 months and median OS of 21 months. Although 43% of patients experienced cytokine release syndrome (grade not specified), only one patient developed immune effector cell-associated neurotoxicity syndrome (ICANS), indicating a manageable safety profile suitable for outpatient and community settings.

These two abstracts underscore the rapid emergence of BsAbs as powerful, targeted options that offer hope for patients with limited alternatives. As ongoing research continues to refine their use, it is increasingly clear that BsAbs will play an integral role in the evolving management strategies of MCL, providing tailored, effective therapies with manageable toxicity profiles for high-risk patient populations.

Visit the SOHO 2025 meeting news page for more coverage from the meeting.