Next Article in Journal
Breed Varieties of Pigs for Disease Resistance and Susceptibility to Seneca Valley Virus Infection
Previous Article in Journal
Major Common Hallmarks and Potential Epigenetic Drivers of Wound Chronicity and Recurrence: Hypothesis and Reflections
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Richter Transformation in Chronic Lymphocytic Leukemia: Current Treatment Challenges and Evolving Therapies

1
Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
2
Department of Nephrology, Kidney Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
3
College of Medicine, Chang Gung University, Taoyuan 333423, Taiwan
4
Division of Hematology/Oncology, Department of Pediatrics, Chang Gung Memorial Hospital in Linkou, Taoyuan 333423, Taiwan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(17), 8747; https://doi.org/10.3390/ijms26178747
Submission received: 20 July 2025 / Revised: 1 September 2025 / Accepted: 3 September 2025 / Published: 8 September 2025

Abstract

Richter transformation (RT) affects 2–10% of chronic lymphocytic leukemia (CLL) patients, evolving into an aggressive lymphoma—most often diffuse large B-cell lymphoma—with poor prognosis, especially when clonally related to CLL. Key risk factors include unmutated IGHV, TP53 and NOTCH1 mutations, stereotyped B-cell receptors, and complex cytogenetics. This review summarizes RT biology, clinical predictors, and treatment outcomes. Traditional chemoimmunotherapy (e.g., R-CHOP) yields complete response rates around 20–30% and median overall survival of 6–12 months; intensified regimens (R-EPOCH, hyper-CVAD) offer only modest gains. Allogeneic hematopoietic stem cell transplantation is potentially curative but limited to fit patients due to high treatment-related mortality. Emerging therapies now include Bruton’s tyrosine kinase and BCL-2 inhibitors, which achieve partial responses but short progression-free survival. CD19-directed chimeric antigen receptor T-cell therapies produce overall response rates of 60–65%, though relapses remain frequent. Bispecific antibodies (e.g., CD3×CD20 agents epcoritamab and mosunetuzumab) show promising activity and tolerable toxicity in relapsed/refractory RT. Ongoing trials are exploring combinations with checkpoint inhibitors, triplet regimens, and novel targets such as ROR1, CD47, and CDK9. Continued research into optimized induction, consolidation, and innovative immunotherapies is essential to improve outcomes in this biologically distinct, high-risk CLL-related lymphoma.

Graphical Abstract

1. Introduction

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in western countries, with an age-adjusted incidence of 4.6 per 1,000,000 individuals [1,2]. Richter transformation (RT) developed in 2 to 15% of CLL patients, with an incidence of 0.5 to 1% per year [3,4]. RT was first described by Maurice Richter in 1928, refers to the evolution of CLL into an aggressive lymphoma, most often diffuse large B-cell lymphoma (DLBCL), with occasional cases of Hodgkin lymphoma or rarer variants like B-lymphoblastic lymphoma and mantle cell lymphoma [5,6,7,8]. RT is characterized by rapidly progressive lymphadenopathies, the presence of B symptoms, elevated lactate dehydrogenase level, and frequent extranodal involvement [3]. The prognosis of RT is generally poor, particularly in cases of clonally related RT, which accounts for approximately 80 percent of all cases [9].
RT is predisposed by both clinical and biological high-risk features: lymphadenopathy > 5 cm, Rai stage III–IV, unmutated immunoglobulin heavy chain variable region (IGHV) status, TP53 aberrations, NOTCH1 mutations, presence of stereotyped B-cell receptors, over-expression of CD38 or ZAP-70, and a complex karyotype [10,11,12,13,14,15,16,17], commonly defined as the presence of ≥3 numeric or structural chromosomal abnormalities by conventional karyotyping [18] A Danish population study confirmed these features at scale, with an RT incidence of 4.5% and a higher risk in early-stage patients who later required treatment [19]. The median overall survival (OS) after a diagnosis of RT-DLBCL remains poor, at approximately 12 months [6]. Given this dismal prognosis, a deeper understanding of the biology and therapeutic landscape of RT is essential. This review aims to explore the molecular pathogenesis, clinical characteristics, and treatment outcomes of RT under various therapeutic strategies, including allogeneic hematopoietic stem cell transplantation (allo-HSCT), chimeric antigen receptor (CAR) T-cell therapy, bispecific antibody therapy, and ongoing or future clinical trials.

2. Molecular Features and Pathogenesis

RT can arise either from the original CLL clone, referred to as clonally related RT, or from a distinct B-cell clone, known as clonally unrelated RT [9,20]. Approximately 80% of RT-DLBCL cases are clonally related, retaining the same IGHV rearrangements as the CLL and reflecting true clonal evolution driven by the stepwise acquisition of high-risk genetic lesions [21]. In contrast, roughly 20% of cases are clonally unrelated, arising as de novo lymphomas that lack a clonal connection to the CLL and typically exhibit clinical behavior closer to sporadic DLBCL, with comparatively superior outcomes [22]. Longitudinal studies of paired CLL and RT samples have illustrated this evolutionary process: Nadeu et al. demonstrated that minor subclones harboring RT-associated mutations can be present years—sometimes nearly two decades—before overt transformation [23]. These latent high-risk subclones may subsequently expand into the dominant RT clone when selective pressures such as prior therapy or microenvironmental cues confer a survival advantage. Over time, multiple competing subclones may diversify in parallel, but ultimately one lineage accumulates a critical constellation of driver lesions and emerges as the dominant aggressive clone that clinically defines RT [23]. This “early seeding” model underscores that RT is not a sudden event but rather the culmination of long-standing clonal diversification and Darwinian competition, highlighting the evolutionary continuum from indolent CLL to aggressive lymphoma.
At molecular level, RT is underpinned by recurrent genetic alterations that disrupt tumor suppressor networks and deregulate key signaling pathways, thereby propelling the indolent CLL clone into a high-grade malignant state. Four lesions in particular—TP53 inactivation, CDKN2A/B loss, NOTCH1 activation, and MYC dysregulation—consistently emerge as central hallmarks of RT pathogenesis, forming a genetic foundation upon which indolent CLL evolves into aggressive lymphoma [21,24]. Importantly, each of these alterations not only drives malignant progression directly but also cooperated with aberrant B-cell receptor (BCR) signaling, a central axis of CLL biology and a critical force in the pathogenesis of RT [24]. Figure 1 schematically illustrates the clonal evolution from CLL to RT and associated therapeutic implications.
Disruption of the TP53 pathway is the most common event in RT-DLBCL, observed in approximately 60–70% of cases [21,24]. TP53 loss, through mutation or deletion of chromosome 17p, disables the DNA damage checkpoint and apoptotic machinery, enabling unchecked accumulation of genomic lesions. Importantly, TP53 mutations are often detectable in the CLL clone prior to transformation, suggesting that TP53-deficient subclones are predisposed to evolve into RT [23]. The functional interaction between TP53 loss and BCR signaling is particularly revealing. In CLL cells with intact checkpoints, BCR stimulation paradoxically induces both pro-proliferative signals and the expression of CDK inhibitors such as p21 and p16, leading to cell-cycle arrest in the absence of co-stimulatory cues. However, when TP53 and CDKN2A/B are inactivated, these braking mechanisms are removed, converting BCR engagement from an anergic to a proliferative signal [25]. Experimental models confirm that CLL cells with combined TP53/CDKN2A inactivation proliferative autonomously upon BCR stimulation and that this proliferation remains strictly BCR-dependent. Thus, TP53 loss not only destabilized the genome but also rewires BCR signals into an engine of unrestrained clonal expansion.
Alongside TP53, deletion in the CDKN2A/B locus (chromosome 9p21) are observed in 20 to 30% RT cases [24]. These genes encode inhibitors of CDK4/6, which regulate the G1-S cell-cycle checkpoint. Their synergy with BCR signaling is profound: in the context of CDKN2A/B deletion, antigen-driven BCR activation directly propels cell-cycle entry without the counterbalance of inhibitory regulators. Functionally, this explained why RT cases with dual TP53 and CDKN2A/B loss are particularly aggressive yet still reliant on BCR-derived signals, reflecting a paradoxical dependency even in highly genomically unstable clones [25]. This cooperative biology highlights the intersection of cell-cycle deregulation and chronic antigenic stimulation in RT pathogenesis.
Oncogenic driver mutations further cooperate with these tumor suppressor losses to drive the clonal evolution of RT. Activating mutations in NOTCH1, typically truncating mutations in the PEST domain, are present in 20 to 30% of RT-DLBCL [21]. These mutations result in persistent NOTCH1 signaling and transcription of survival-promoting genes. Clinically, NOTCH1 mutations are a well-established risk factor for RT: up to 45% of CLL patients harboring NOTCH1 mutations eventually transform [24]. Importantly, NOTCH1 signaling directly amplifies BCR downstream cascades, particularly PI3K-AKT and NF-κB pathways, promoting proliferation and apoptosis resistance. This convergence fosters a pro-survival transcriptional program, while also reducing reliance on external microenvironmental support. NOTCH1 mutations are more frequent in CLL patients on BTK inhibitors, suggesting that therapeutic blockade of proximal BCR kinases may select for clones in which NOTCH1 substitutes as an alternative amplifier of PI3K/AKT signaling. Thus, NOTCH1 lesion exemplifies how RT biology adapts by sustaining oncogenic BCR outputs through parallel pathways [22].
Another major driver is dysregulation of MYC, a master oncogene that controls cellular proliferation and metabolism. MYC abnormalities are identified in 30 to 50% of RT cases, occurring via various mechanisms including gene amplifications, chromosomal translocations, or loss-of-function mutations in negative regulators such as MGA [24]. MYC activation drives metabolic reprogramming, cell-cycle acceleration, and transcriptional programs favoring aggressive growth. In RT, MYC alterations often co-occur with TP53 inactivation, producing highly proliferative and genomically unstable clones. This synergy is thought to underline the explosive clinical behavior of RT-DLBCL. MYC activity also integrates with BCR signaling: NF-κB and PI3K/AKT cascades downstream of BCR upregulate MYC expression, while MYC in turn enhances glycolysis and oxidative phosphorylation, meeting the energetic demands of chronic signaling. Transcriptomic profiling of RT cells demonstrates enrichment of oxidative phosphorylation programs compared with parent CLL, consistent with MYC-driven metabolic rewiring layered upon BCR stimulation. This cooperation explains why MYC-aberrant RT clones often dominate the evolutionary landscape—they are not only genetically unstable but metabolically optimized for survival in the context of chronic antigenic signaling.
Beyond single-gene lesions, RT is distinguished by profound genomic instability. Whole-genome and exome sequencing have revealed RT harbors significantly more mutations, copy number alterations, and structural rearrangements than the antecedent CLL [21]. Catastrophic genomic events such as chromothripsis and chromoplexy are frequent, and around 15% of RT-DLBCLs exhibit whole-genome doubling, a phenomenon rarely seen in untreated CLL [21]. These phenomena reflect both the disabling of DNA damage checkpoints (via TP53/ATM defects) and the proliferative drive imposed by BCR and MYC. Intrinsic processes such as aberrant somatic hypermutation and telomere erosion further fuel mutagenesis, while extrinsic factors—including therapy-induced stress and microenvironmental signals—likely accelerate genomic chaos [24]. Ultimately, Darwinian selection among these genomically diverse subclones yields one dominant lineage bearing the optimal constellation of BCR signaling capacity, checkpoint disruption, and proliferative advantage.
The immunogenetic context of BCR signaling further supports its role in RT. Clonally related RT frequently arises from CLL clones with unmutated IGHV genes and stereotyped BCRs, particularly subset #8, which recognizes autoantigens such as vimentin and delivers strong signaling outputs [21]. Such clones are strongly enriched among patients who progress to RT, highlighting the contribution of chronic antigenic drive. High ZAP-70 expression in these CLL cells lowers the threshold for BCR activation, enhancing downstream cascades. In contrast, clonally unrelated RT, which resembles de novo germinal-center DLBCL, typically arises from mutated IGHV cells and lacks stereotyped BCRs, underscoring distinct pathogenic trajectories.
Morphologically, RT-DLBCL predominantly exhibits an activated B-cell or non-germinal center phenotype, consistent with its dependence on chronic active BCR signaling. Many cases retain CLL immunophenotypic features, including CD5 expression. Compared with de novo DLBCL, RT more frequently expresses PD-1/PD-L1, facilitating immune evasion. Integrative analyses now define molecular subtypes of RT based on driver combinations: NOTCH1/trisomy 12 and SF3B1/EGR2-mutant subtypes show somewhat lower genomic complexity, while TP53-deficient clones with whole-genome doubling display the greatest instability and poorest outcomes [21]. However, these proposed classifications are based on limited cohorts, and their reproducibility across diverse populations remains uncertain.
Despite advances, significant challenges remain in delineating RT pathogenesis. Most genomic studies have focused on heavily pretreated CLL, raising questions about whether therapy accelerates particular evolutionary paths. The temporal order of driver lesions and their relative contributions to transformation versus general disease progression remain incompletely defined, given the scarcity of prospective longitudinal data. Furthermore, while BCR signaling clearly plays a central role, the extent to which transformation is driven by intrinsic antigenic stimulation versus microenvironmental support is not fully resolved. Finally, functional insights largely stem from in vitro or murine models, which cannot fully recapitulate human CLL evolution or the stromal context of transformation. Improved longitudinal sampling, single-cell multi-omics, and patient-derived models will be critical to address these gaps.
In summary, RT represents the endpoint of prolonged clonal evolution in CLL, characterized by interplay between chronic BCR signaling and recurrent driver lesions. TP53 inactivation, CDKN2A/B loss, NOTCH1 activation, and MYC dysregulation converge with hyperactive BCR pathways to dismantle tumor-suppressor programs, remodel metabolism, and promote genomic instability. These synergistic events forge the dominant lymphoma clone, culminating in the aggressive and often treatment-refractory biology of RT.

3. RT Incidence and Outcomes

High-risk genomic alterations in CLL, including TP53 aberrations, NOTCH1 mutations, complex karyotype, and unmutated IGHV, are consistently associated with increased RT risk [10,11,12,13,14,15,16,17]. These factors, together with clinical features and prior therapy, support risk-adapted treatment planning and trial referral in RT [3,26].
Reported RT incidence varies widely depending on treatment era, patient population, diagnostic criteria, and follow-up duration. A summary of major studies and cohorts is shown in Table 1.
Differences in incidence likely reflect patient selection, biopsy practices, treatment exposure, and biological effects of therapy. Hypothesized mechanisms include selection of pre-existing aggressive subclones under therapeutic pressure, therapy-induced immune dysfunction, and microenvironmental changes that facilitate transformation. For example, prolonged BTK inhibition may promote outgrowth of TP53-mutated clones, while venetoclax may preferentially spare apoptosis-resistant populations.
Prospective data on RT are limited. A German CLL Study Group pooled analysis of frontline chemoimmunotherapy (CIT) trials reported RT in 3% of patients, with median OS of 9.4 months post-RT [28]. Earlier cohorts from MD Anderson and Mayo Clinic showed median OS of 8 and 7.8 months, respectively [6,27]. However, RT diagnosed in previously untreated CLL patients showed better outcomes with median survival up to 46.3 months, suggesting that treatment-naïve status at the time of transformation may be linked to less adverse outcomes [6,9].
Real-world registry data and clinical trial data support these findings. A Danish study found RT risk unchanged in the era of novel agents, with chemotherapy exposure linked to worse survival [19]. In RESONATE and RESONATE-2 trials, 4 to 5% of patients developed RT after ibrutinib [30,31]. Similar observations are reported in trials of venetoclax [32]. The Bruton’s tyrosine kinase inhibitor (BTKi) era brought hope of altering RT incidence and treatment. Frontline ibrutinib may reduce RT risk to 1 to 4% annually, but when progression occurs, it is often presented as RT [19,33,34]. Analyses from Ohio State and MD Anderson revealed that 30 to 40% of patients progressing on ibrutinib developed RT, with a dismal median OS of 3 to 6 months [34,35]. Collectively, these data suggest that although novel agents can delay CLL progression, they do not eliminate the risk of RT.
Notably, emerging trials such as STELLAR now stratify patients according to treatment history, distinguishing between treatment-naïve and previously treated populations, in recognition of their differing prognosis [36]. This trial is scheduled for completion in 2027 and reflects a broader shift toward personalized and risk-adapted therapeutic strategies in RT.
MRD is a validated prognostic tool in CLL, typically measured by multicolor flow cytometry (~10−4 sensitivity) or next-generation sequencing (~10−6) [37,38]. Its prognostic role in RT remains undefined and should be interpreted cautiously. Recent CAR T-cell therapy studies in RT report high response rates but have not consistently integrated MRD assessment into outcome measures CLL [39,40]. While MRD negativity pre-allo-HSCT correlates with superior outcomes in aggressive lymphoma cohorts, specific data in RT are lacking [40,41]. Prospective RT trials incorporating MRD as a biomarker to guide consolidation strategies would be worthwhile.

4. Treatment Outcomes Across CIT, BTKi, and BCL2 Inhibitor Eras

Multiple treatment strategies are now available for managing RT (Table 2). In this review, response definitions are based on the Lugano 2014 criteria for lymphoma. Complete response (CR) is characterized by Positron Emission Tomography/Computed Tomography (PET/CT) negativity and complete resolution of disease. Partial response is defined as a reduction of at least 50% in measurable disease without evidence of new lesions. Relapse denotes the presence of new or progressive diseases. MRD refers to subclinical disease identified through flow cytometry or molecular methods [42]. Historically, RT was treated as de novo DLBCL using conventional CIT regimens such as R-CHOP [42]. However, outcomes have been poor, with CR rates of only around 20% and median OS ranging from 6 to 12 months [6,43]. Attempts to improve efficacy through intensified regimens, such as hyper-CVAD or platinum-based combinations, have yielded only modest improvements in response rates without meaningful extension of OS [27,44,45,46,47]. For example, dose-adjusted R-EPOCH has demonstrated response rates as high as 67% in some series; however, its benefit is limited by early relapse, particularly among patients with complex karyotypes [48]. The French Richter study similarly reported an overall response rate (ORR) of 60% using platinum-based CIT, yet 2-year survival without transplant remained under 20% [27,44].
BTKi used as monotherapy in established RT have shown limited efficacy. Ibrutinib and acalabrutinib yield ORRs of 38 to 40% with median progression-free survival (PFS) of 3 to 4 months and few durable responses [49,50,51]. Zanubrutinib plus the PD-1 inhibitor tislelizumab modestly improved ORR to 58% in a phase II study [52]. Non-covalent BTKi such as pirtobrutinib have also demonstrated encouraging activity, with an ORR of 50% and a CR rate of 13%; however, the median PFS remained limited at 3.7 months [53]. These findings suggest that while BTKi may provide partial disease control in RT, they are rarely sufficient as monotherapy.
Venetoclax, a BCL2 inhibitor, has shown promise in RT, especially in combination. Although RT can develop during venetoclax treatment, often early in the clinical course, responses to venetoclax monotherapy are minimal, with a reported median OS of only 1.1 months in affected patients [54,55]. In contrast, combining venetoclax with CIT has yielded more. In a phase I study, the combination of venetoclax with dose-adjusted R-EPOCH (VR-EPOCH) achieved an ORR of 90% and a CR rate of 53% [56]. A multicenter phase II trial evaluating venetoclax plus R-CHOP (VR-CHOP) reported an ORR of 65% and a CR rate of 46% [57]. Retrospective analyses further support the efficacy of venetoclax-based combinations, particularly when paired with CIT, with ORR up to 55% and some patients achieving durable remissions following allo-HSCT [58]. The MOLTO trial investigated a chemotherapy-free regimen combining venetoclax, obinutuzumab, and the PD-L1 inhibitor atezolizumab, which resulted in an ORR of 67.9% and a CR rate of 28.6%, demonstrating that deep and sustained responses are achievable without cytotoxic chemotherapy [59,60]. Additional ongoing studies are evaluating venetoclax in combination with duvelisib or as part of triplet regimens with BTKi and anti-CD20 antibodies, aiming to further improve treatment outcomes in RT [61,62]. Despite encouraging results from combination regimens, BTKi monotherapy in RT generally yields only modest ORR and median PFS of 3 to 4 months, with few durable remissions [49,50,51]. Venetoclax monotherapy has shown limited efficacy, with some series reporting median OS as short as 1.1 months in affected patients [54,55]. It is important to note that most studies evaluating CIT, BTKi, and BCL2 inhibitor–based regimens in RT are retrospective or early-phase single-arm trials. These designs are inherently prone to selection bias, heterogeneous patient populations, and non-standardized response assessments, which limit the ability to directly compare outcomes across studies or to generalize results to all patients with RT.
Altogether, while conventional CIT remains inadequate, venetoclax-based combinations, particularly when used as a bridge to stem cell transplantation or cellular therapies, represent a significant advancement in the treatment of RT.

5. Role of Allo-HSCT

Allo-HSCT remains the only potentially curative treatment option for a subset of patients with RT [63], although it is associated with both significant benefits and notable limitations. Given the limited durability of response to CIT and novel agents, allo-HSCT is typically considered as a consolidative strategy for eligible patients who achieve remission after induction therapy [64,65,66]. In practice, allo-HSCT eligibility for RT typically requires Eastern Cooperative Oncology Group performance status 0–2, preserved organ function, and controlled infection. Conditioning regimens are often adjusted based on age, with reduced-intensity protocols offered to older patients to mitigate toxicity. Pre-transplant comorbidities are commonly assessed using the Hematopoietic Cell Transplantation–Specific Comorbidity Index, which has been shown to predict treatment-related mortality (TRM) and OS [67,68,69,70]. These patient- and disease-related factors, along with disease status at transplant, donor availability, and patient preferences, explain why only a subset of RT patients proceed to allo-HSCT.
The depth of response prior to allo-HSCT is a critical prognostic factor in RT. Prior study has shown that patient who undergo transplant in a responsive disease status have better outcomes than those transplanted with active disease [27]. The essential pre-transplant response parameters are achieving a radiographic CR on PET/CT (no sites of active uptake) and clearance of disease on biopsy (bone marrow and any previously involved nodes)—essentially, a CR by Lugano 2014 criteria. Patients in CR have significantly better transplant outcomes [71]. Partial responders can still proceed to transplant if that is the best response achievable, but their post-transplant outcomes are less optimal than those of patients in CR. In contrast, neither guidelines nor studies currently require molecular remission that achieved negative MRD before transplant, as its prognostic significance in RT remains undefined. The priority is achieving a clinical and metabolic remission, which has demonstrable impact on improving post-transplant survival in patients with RT.
Although no randomized trials have been conducted, multiple retrospective studies have demonstrated that long-term survival is possible after transplant in RT. In the Mayo Clinic series, 24 of 204 RT patients underwent transplantation (20 autologous, 4 allogeneic); the median OS post-transplant was 55.4 months, significantly longer than the approximately 12-month median OS in non-transplanted patients [6]. However, these results should be interpreted with caution, as transplant recipients were highly selected individuals who were younger, had fewer comorbidities, and were in remission at the time of transplant.
The Center for International Blood and Marrow Transplant Research reported outcomes from a multi-center cohort, showing a 3-year OS of 43 to 61% and PFS of 34 to 52% after allo-HSCT [71]. In contrast, autologous-HSCT offered slightly better short-term outcomes due to lower TRM. However, the tradeoff was a higher relapse rate and limited long-term survival. The curative potential of allo-HSCT is largely attributed to the graft-versus-lymphoma effect, although it carries significant high risks. TRM remains substantial, with early transplant-related deaths approaching 25% in the study [72]. In a retrospective study conducted by the European Group for Blood and Marrow Transplantation, among 59 patients with RT, approximately half received allo-HSCT. The 2-year OS was 39% for allogeneic and 36% for autologous HSCT [41,73]. Notably, only patients receiving allogeneic grafts achieved sustained remissions beyond 3 years, further supporting allo-HSCT as the preferred curative strategy. These findings suggest that although autologous-HSCT may be considered for patients with chemosensitive disease who are not eligible for allo-HSCT, it is less likely to achieve durable remission due to the persistent risk of relapses from residual CLL or transformed clones. Therefore, autologous-HSCT should not be regarded as a first-line curative option.
Despite these promising data, many patients with RT are older and have comorbidities due to prior CLL treatment, limiting their eligibility for myeloablative conditioning. Reduced-intensity conditioning (RIC) allo-HSCT regimens have been explored as an alternative. For example, an MD Anderson series using RIC in 8 patients with RT reported that 3 remained alive and disease-free at over 3 years [73,74]. A French multicenter study of 17 patients reported a 2-year OS of 47%, though relapse and non-relapse mortality remained high [75].
An “intention-to-treat” analysis from Canada highlighted the challenge of reaching transplant: many patients referred never underwent transplantation due to rapid disease progression or fitness decline [72]. This underscores the need for early referral and effective induction strategies to enable timely transplant.
Allo-HSCT remains the only potentially curative therapy for RT; however, relapses following transplant is a concern, prompting interest in post-transplant maintenance strategies. Given the rarity and heterogeneity of RT, maintenance therapy after allo-HSCT remains an area with very limited evidence. Donor lymphocyte infusion for early relapses is the one strategy with some documented success [76,77]. A recent multicenter study by Eikema et al. briefly discussed donor lymphocyte infusion and maintenance therapies in high-risk RT patients undergoing allo-HSCT, although no standardized maintenance regimen was uniformly employed [41]. Targeted agents, including venetoclax, ibrutinib, and checkpoint inhibitors, have not been validated as post-transplant maintenance therapies, and their routine use is not supported in the absence of trial data.
However, allo-HSCT is associated with substantial early and late toxicities, including acute and chronic graft-versus-host disease, organ toxicities, prolonged cytopenias, and treatment-related mortality rates up to 20 to 25%. Long-term survivors often experience moderate to severe late complications affecting multiple organ systems, with fatigue and insomnia remaining common [78].
Overall, allo-HSCT provides the best chance of long-term remission or cure in RT, especially for younger, fit patients who achieve CR. Early transplant consultation should be considered in all eligible patients with RT responding to initial therapy. Autologous transplantation may postpone relapses in certain cases, though relapses occur frequently and long-term remissions are uncommon. The available allo-HSCT data for RT are derived almost entirely from retrospective series with small sample sizes and significant selection bias toward younger, fitter patients who achieve remission before transplant. The absence of prospective randomized studies means that reported survival benefits must be interpreted with caution, and outcomes may not be replicable in broader clinical practice. Future directions include improving induction regimens to enable more patients to reach transplant, exploring maintenance strategies (e.g., post-alloHSCT BTKi or BCL2 inhibitor), and evaluating CAR T-cell therapy as a consolidative or alternative approach.

6. CAR T-Cell Therapy in RT

Anti-CD19 CAR T-cell therapy has revolutionized the management of relapsed or refractory DLBCL (r/r DLBCL). Given that RT often presents as an aggressive, CD19-positive lymphoma, there has been substantial interest in applying CAR T-cell therapy to this setting. However, the application of CAR-T cell therapy in patients with RT involves several practical considerations (Table 3).
In the previous pivotal clinical trial of anti-CD19 CAR T-cell therapy for r/r DLBCL, patients with RT were not included because RT is regarded as a distinct disease entity [79,80]. While no CAR T-cell product is specifically approved for RT, several retrospective studies and clinical trials have evaluated its efficacy, largely through off-label use (Table 4).
In CLL, the CR rate for lisocabtagene maraleucel (liso-cel) was 18%, which is lower than the rate observed in DLBCL, and infusion products often show high levels of T-cell exhaustion [87,88,89]. This raises the concern that CAR T-cell therapy may have reduced efficacy in RT compared to DLBCL. RT differs immunologically from CLL, showing high PD1 expression and strong responses to PD1 inhibitors [52,90,91], while CLL shows minimal response to PD1 inhibition [92].
An international multicenter retrospective study assessed 69 patients with RT treated with commercial anti-CD19 CAR T-cell therapies—axicabtagene ciloleucel, tisagenlecleucel, or liso-cel—following extensive prior therapy, including BTKi and BCL-2 inhibitors in 84% of cases [82]. The ORR was 63%, with a CR rate of 46%. Those who achieved CR experienced durable responses, with a median duration of response (DoR) of 27.6 months. Of note, detectable CLL MRD was eradicated in 81% of patients. A 2025 multicenter analysis confirmed durable remissions in a subset of patients, though relapse remains common, especially in TP53-aberrant cases [93].
Two additional retrospective studies have corroborated that CAR T-cell therapy for RT yields response rates comparable to those observed in de novo DLBCL. One study compared outcomes between patients with RT (n = 30) and those with aggressive B-cell lymphoma (n = 283) undergoing CAR T-cell therapy, finding similar safety profiles but reduced efficacy in the RT group [81]. At day 100, the ORR and CR in RT patients were 57% and 47%, respectively. Furthermore, RT patients exhibited inferior median (9.9 vs. 18 months) and 12-month OS rates (45% vs. 62%) compared to DLBCL, with worse survival associated with higher numbers of prior therapies, elevated LDH, and RT histology. In another international retrospective analysis conducted by the European research initiative on CLL, an ORR of 65% and a CR rate of 50% were reported among 54 RT patients receiving CAR T-cell therapy [83].
CAR T-cell therapy is associated with notable acute toxicities, including cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), which may present with confusion, language disturbances, and cognitive deficits [94]. Notably, even in a predominantly older and heavily pretreated population, toxicity rates were manageable, with grade ≥3 CRS and ICANS occurring at frequencies comparable to those observed in patients with transformed indolent non-Hodgkin lymphoma and de novo DLBCL [81]. Long-term follow-up data indicate that approximately 38 to 50% of survivors experience cognitive difficulties, anxiety, or depression [94,95,96]. Qualitative studies also report persistent impairments in domains such as sleep, role functioning, and emotional well-being, underscoring the importance of comprehensive survivorship care [97].
These initial response rates are similar to those seen in pivotal trials for r/r de novo DLBCL [79,80,98], indicating that RT cells remain sensitive to CAR T-cell cytotoxicity. However, response durability is lower; two retrospective studies with 2-year median follow-up reported median PFS of 4.7 and 4.3 months, and median OS of 8.5 and 9.9 months, respectively [81,82]. These outcomes are notably poorer than those observed in prior pivotal trials of anti-CD19 CAR T-cell therapy for de novo DLBCL [80,98,99].
The reasons for reduced durability in RT remain under investigation. Potential contributing factors include a highly immunosuppressive CLL microenvironment that may impair CAR T-cell persistence, higher tumor burden at the time of infusion, and the expression of checkpoint ligands or loss of target antigen (CD19) in RT cells [40,81]. To address the limitations associated with response durability, post-CAR T-cell consolidation strategies—such as allo-HSCT—may improve long-term outcomes in selected patients, warranting further investigation. Additional approaches, including combination therapies, are currently under active exploration. Notably, two early-phase clinical trials have attracted attention. One trial is evaluating the concurrent administration of a PD-1 checkpoint inhibitor (nivolumab) and a BTKi (ibrutinib) around the time of CAR T-cell infusion, based on the premise that ibrutinib may enhance T-cell fitness while nivolumab mitigates PD-1–mediated suppression [90]. Another trial is investigating CAR T-cell therapy in combination with zanubrutinib, a next-generation BTKi, in cases of relapsed RT (NCT05873712). These studies aim to overcome tumor microenvironment barriers and promote sustained CAR T-cell persistence.
Furthermore, next-generation therapeutic strategies are in development, including CAR T-cell constructs targeting alternative antigens in response to CD19 loss observed in certain RT cases, as well as modified cell therapies engineered to resist exhaustion or evade immunosuppressive mechanisms.
In summary, CD19 CAR T-cell therapy offers a promising but still limited option for patients with RT. The response rates are comparable to those in de novo DLBCL, but the majority of RT patients relapse within the first year, resulting in modest improvement of median PFS and OS. Nevertheless, CAR T-cell therapy remains an important treatment avenue for patients who are ineligible for allo-HSCT and have exhausted chemotherapy and novel agents. It may also serve as a bridge to transplant or long-term remission in a subset of patients. While CAR T-cell therapy achieves high initial responses, two large retrospective series reported median PFS of only 4.3 to 4.7 months and median OS less than 10 months [81,82], highlighting the substantial risk of early relapse even after remission. Published CAR T-cell therapy data in RT primarily originates from retrospective, multicenter registries or small institutional experiences. These analyses often lack standardized eligibility criteria, uniform response assessment, and consistent follow-up durations, which may overestimate efficacy or underestimate late toxicities. Ongoing research into patient selection, optimal timing, and adjunctive therapies will be essential to fully realize the potential of CAR T-cell therapy in RT.

7. Bispecific Antibody Therapies

Bispecific T-cell engaging antibodies represent a novel and promising class of immunotherapy for B-cell malignancies, including RT. These agents function by simultaneously binding CD3 on T-cells and a B-cell antigen, most commonly CD20, thereby redirecting cytotoxic T-cell activity against malignant B-cells. Therefore, bispecific antibodies serve as a potential alternative, given that patients with CLL often exhibit T-cell dysfunction (Table 5) [100].
Epcoritamab, a subcutaneously administered CD3×CD20 bispecific IgG1 antibody, has demonstrated notable efficacy in LBCL and is now being evaluated in RT. In the ongoing EPCORE CLL-1 phase I/II trial, 35 evaluable RT patients received single-agent epcoritamab [103]. The ORR was approximately 50%, with 35% achieving CR, several of which were also MRD-negative. Median PFS was 12.8 months. Considering that these patients were heavily pretreated, these results are noteworthy. Cytokine release syndrome CRS, mostly grade 1–2, occurred but was manageable with step-up dosing. These results establish epcoritamab as a potent and potentially durable treatment for RT. Combination studies are ongoing to further enhance efficacy.
Mosunetuzumab, another CD3×CD20 bispecific antibody administered intravenously, is already approved for relapsed follicular lymphoma and has shown activity in DLBCL. In a first-in-human study, 20 RT patients received mosunetuzumab monotherapy [104]. Among them, the ORR was 40%, with 20% achieving CR. Some remissions were durable beyond 6 months. Toxicity was similar to other lymphomas, with low-grade CRS and minimal neurotoxicity. Though the CR rate was lower than with epcoritamab, these patients were heavily pretreated, and mosunetuzumab still showed meaningful single-agent activity. An ongoing trial (NCT06521996) is combining mosunetuzumab with CHOP chemotherapy in frontline RT, aiming to improve initial disease control and deepen remission.
Glofitamab, a CD3×CD20 bispecific with a unique 2:1 binding format, is approved in Europe for relapsed/refractory DLBCL. While dedicated RT-specific data are limited, case reports suggest clinical responses in RT patients, even inducing CR [100,101]. Ongoing expansion cohorts include RT patients, and the drug is considered a viable option, especially in patients who relapse after CAR T-cell therapy or are ineligible for it. CRS is manageable with standard step-up dosing and obinutuzumab pre-treatment.
Ongoing studies such as Euplagia-1 are testing dual-targeting CAR T and immune engagers with promising early activity [104]. Trials of blinatumomab, a CD19-directed BiTE, have reported encouraging results in RT, including durable MRD-negative responses in selected patients [105].
In summary, bispecific antibodies offer an important new therapeutic class in RT. Their key advantages include off-the-shelf availability, repeat dosing potential, and relatively manageable toxicity profiles. Epcoritamab in particular has shown promising response rates, including MRD-negative CR. As more data emerge, these agents may be positioned in multiple treatment lines—either as monotherapy in relapsed RT, in combination regimens, or following CAR T-cell failure. Future trials will determine whether these therapies can serve as curative platforms, particularly when paired with consolidation strategies. Bispecific antibodies are poised to become a cornerstone in the evolving management of RT.

8. Ongoing and Future Clinical Trials

The urgent need for more effective therapies in RT has spurred a wave of clinical trials exploring novel strategies (Figure 2). These include combination regimens, immunotherapies, and next-generation cellular and targeted approaches in both frontline and relapsed settings. Below is a summary of key ongoing or planned trials and future directions.

8.1. Combination Chemo-Immunotherapy Trials

In addition to the STELLAR study, which compares standard R-CHOP with R-CHOP plus acalabrutinib [36], another ongoing trial (NCT04679012) is evaluating polatuzumab vedotin—an anti-CD79b antibody-drug conjugate—in combination with R-CHOP. Since CD79b is frequently expressed in RT cells, the addition of polatuzumab may potentiate R-CHOP efficacy. Preliminary results will guide further development of this chemo-immunotherapy strategy.

8.2. Checkpoint Inhibitor Combinations

Recent data suggest a potential role for checkpoint blockade in RT. A phase II study of pembrolizumab demonstrated responses in heavily pretreated RT patients, particularly those with prior BTKi exposure [106]. Copanlisib plus nivolumab also showed activity, with manageable toxicity, in a multicenter trial [107]. Ongoing work also includes duvelisib with nivolumab (NCT03892044), based on the hypothesis that PI3K inhibition may modulate the tumor microenvironment to enhance T-cell responses. Early data from a phase II trial also suggest that combining pembrolizumab with BTKi may improve outcomes compared to PD-1 monotherapy [108].

8.3. Triple Targeted Therapy Regimens

“Time-limited” triplet therapies are gaining traction. Trial NCT05536349 investigates pirtobrutinib (non-covalent BTKi), venetoclax, and obinutuzumab in newly diagnosed CLL and RT. This trial’s inclusion of RT in a frontline CLL cohort is novel and may illuminate strategies to intercept early transformation. Another trial, DTRM-555, evaluates a triplet of investigational BTKi DTRMWXHS-12, everolimus (mTOR inhibitor), and pomalidomide (an immunomodulator) [109]. Preliminary findings indicate certain responses, but myelosuppression remains a potential issue. Not all investigational regimens have been successful. The phase II study was discontinued in December 2024 due to financial constraints [110]. This underscores the challenges of developing effective therapies in a rare and heterogeneous disease. These studies test whether targeting multiple oncogenic pathways simultaneously can suppress RT escape mechanisms.

8.4. CAR T-Cell Therapy Enhancements

New strategies seek to improve CAR T-cell efficacy in RT. One NCI-sponsored trial (NCI-2022-10247) combines nivolumab and ibrutinib with CD19 CAR T-cells to enhance expansion and persistence. Another study (NCI-2023-03669) pairs zanubrutinib with CD19 CAR T therapy. Exploratory platforms using “armored” CAR T-cells, which are engineered to express cytokines or PD-1 dominant negative receptors, are being developed specifically for RT. International consortium efforts are also collecting real-world data to refine CAR T-cell use in this population [111].

8.5. Bispecific Antibodies and Emerging Immunotherapies

Building on early success, bispecific antibodies are moving into combination trials. Epcoritamab is being evaluated in the ongoing EPCORE CLL-1 trial, with expansion cohorts testing it in RT and planned combinations with BTKi or venetoclax (NCT04623541). Mosunetuzumab is being combined with CHOP in the frontline RT setting [111]. Additionally, immunotherapies targeting macrophage checkpoints like CD47 are being studied. Evorpacept (ALX148), a CD47-blocking agent, showed early signs of activity in aggressive lymphomas and may be explored in RT [112]. Novel immune cell engagers, such as NK-cell or next-gen T-cell engagers targeting CD19, ROR1, and others, are under preclinical evaluation [113,114,115].

8.6. Targeting ROR1 and Novel Molecular Pathways

ROR1, an embryonic surface protein, is highly expressed in CLL and may persist in RT. A novel antibody-drug conjugate, zilovertamab vedotin, targets ROR1 and has shown activity in mantle cell lymphoma and CLL [116]. The planned waveLINE-006 phase 2 trial will test this agent in aggressive B-cell lymphomas, potentially including RT. Other pathways under investigation include MYC, BCL6, and CDK9. For instance, selective CDK9 inhibitors are being tested to overcome venetoclax resistance by downregulating MCL1 [115]. Molecular profiling of RT biopsies continues to search for targetable mutations unique to RT, though most mutations currently overlap with high-risk CLL.

9. Conclusions

RT remains highly aggressive and treatment-resistant, with limited long-term survival despite recent therapeutic advances. Conventional CIT offers poor durability, while allogeneic stem cell transplantation may provide a potential cure but carries substantial non-relapse mortality, particularly in older or frail patients. Newer approaches have shown promise in select cases, including BTKi and BCL2 inhibitors, CAR T-cell therapy, and bispecific antibodies. Bispecific antibodies like epcoritamab are especially encouraging as off-the-shelf immunotherapies. Ongoing clinical trials are exploring combination strategies and novel targets such as ROR1 and CD47. Continued research and early referral remain critical to improving outcomes in this high-risk population.

Author Contributions

Conceptualization, Z.-C.L. and Y.-J.S.; Literature review, Z.-C.L., M.-J.C. and Y.-J.S.; Manuscript drafting, Z.-C.L., M.-J.C. and Y.-J.S.; Critical revision and editing: M.-J.C., T.-H.J., T.-L.L., Y.-S.H. and Y.-J.S.; Data visualization, Z.-C.L. and Y.-J.S.; Supervision, Y.-J.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CLLChronic lymphocytic leukemia
RTRichter transformation
DLBCLDiffuse large B-cell lymphoma
IGHVImmunoglobulin heavy chain variable region
OSOverall survival
allo-HSCTAllogeneic hematopoietic stem cell transplantation
CARChimeric antigen receptor
CITChemoimmunotherapy
BTKiBruton tyrosine kinase inhibitor
CRComplete response
ORROverall response rate
PFSProgression-free survival
MRDMinimal residual disease
TRMTreatment-related mortality
RICReduced-intensity conditioning
r/r DLBCLRelapsed or refractory diffuse large B cell lymphoma
Liso-celLisocabtagene maraleucel
CRSCytokine release syndrome
ICANSImmune effector cell-associated neurotoxicity syndrome

References

  1. Tsimberidou, A.M.; Wen, S.; O’Brien, S.; McLaughlin, P.; Wierda, W.G.; Ferrajoli, A.; Faderl, S.; Manning, J.; Lerner, S.; Mai, C.V.; et al. Assessment of chronic lymphocytic leukemia and small lymphocytic lymphoma by absolute lymphocyte counts in 2,126 patients: 20 years of experience at the University of Texas M.D. Anderson Cancer Center. J. Clin. Oncol. 2007, 25, 4648–4656. [Google Scholar] [CrossRef]
  2. Hallek, M. Chronic Lymphocytic Leukemia: 2025 Update on the Epidemiology, Pathogenesis, Diagnosis, and Therapy. Am. J. Hematol. 2025, 100, 450–480. [Google Scholar] [CrossRef]
  3. Parikh, S.A.; Kay, N.E.; Shanafelt, T.D. How we treat Richter syndrome. Blood 2014, 123, 1647–1657. [Google Scholar] [CrossRef]
  4. Roberts, A.W.; Davids, M.S.; Pagel, J.M.; Kahl, B.S.; Puvvada, S.D.; Gerecitano, J.F.; Kipps, T.J.; Anderson, M.A.; Brown, J.R.; Gressick, L.; et al. Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2016, 374, 311–322. [Google Scholar] [CrossRef]
  5. Richter, M.N. Generalized Reticular Cell Sarcoma of Lymph Nodes Associated with Lymphatic Leukemia. Am. J. Pathol. 1928, 4, 285–292.7. [Google Scholar]
  6. Wang, Y.; Tschautscher, M.A.; Rabe, K.G.; Call, T.G.; Leis, J.F.; Kenderian, S.S.; Kay, N.E.; Muchtar, E.; Van Dyke, D.L.; Koehler, A.B.; et al. Clinical characteristics and outcomes of Richter transformation: Experience of 204 patients from a single center. Haematologica 2020, 105, 765–773. [Google Scholar]
  7. Pistoia, V.; Roncella, S.; Di Celle, P.F.; Sessarego, M.; Cutrona, G.; Cerruti, G.; Boccaccio, G.P.; Grossi, C.E.; Foà, R.; Ferrarini, M. Emergence of a B-cell lymphoblastic lymphoma in a patient with B-cell chronic lymphocytic leukemia: Evidence for the single-cell origin of the two tumors. Blood 1991, 78, 797–804. [Google Scholar] [CrossRef]
  8. Klener, P.; Fronkova, E.; Berkova, A.; Jaksa, R.; Lhotska, H.; Forsterova, K.; Soukup, J.; Kulvait, V.; Vargova, J.; Fiser, K.; et al. Mantle cell lymphoma-variant Richter syndrome: Detailed molecular-cytogenetic and backtracking analysis reveals slow evolution of a pre-MCL clone in parallel with CLL over several years. Int. J. Cancer 2016, 139, 2252–2260. [Google Scholar] [CrossRef] [PubMed]
  9. Kittai, A.S.; Huang, Y.; Miller, S.; Allan, J.N.; Bhat, S.A.; Bond, D.A.; Brander, D.M.; Byrd, J.C.; Chavez, J.C.; Chong, E.; et al. Outcomes of patients with Richter transformation who received no prior chemoimmunotherapy for their CLL. Blood Cancer J. 2025, 15, 23. [Google Scholar] [CrossRef] [PubMed]
  10. Parikh, S.A.; Rabe, K.G.; Call, T.G.; Zent, C.S.; Habermann, T.M.; Ding, W.; Leis, J.F.; Schwager, S.M.; Hanson, C.A.; Macon, W.R.; et al. Diffuse large B-cell lymphoma (Richter syndrome) in patients with chronic lymphocytic leukaemia (CLL): A cohort study of newly diagnosed patients. Br. J. Haematol. 2013, 162, 774–782. [Google Scholar] [CrossRef] [PubMed]
  11. Ben-Dali, Y.; Hleuhel, M.H.; Andersen, M.A.; Brieghel, C.; Clasen-Linde, E.; Da Cunha-Bang, C.; Niemann, C.U. Risk Factors Associated with Richter’s Transformation in Patients with Chronic Lymphocytic Leukemia. Blood 2018, 132 (Suppl. S1), 1697. [Google Scholar] [CrossRef]
  12. Rossi, D.; Cerri, M.; Capello, D.; Deambrogi, C.; Rossi, F.M.; Zucchetto, A.; De Paoli, L.; Cresta, S.; Rasi, S.; Spina, V.; et al. Biological and clinical risk factors of chronic lymphocytic leukaemia transformation to Richter syndrome. Br. J. Haematol. 2008, 142, 202–215. [Google Scholar] [CrossRef]
  13. Rossi, D.; Rasi, S.; Fabbri, G.; Spina, V.; Fangazio, M.; Forconi, F.; Marasca, R.; Laurenti, L.; Bruscaggin, A.; Cerri, M.; et al. Mutations of NOTCH1 are an independent predictor of survival in chronic lymphocytic leukemia. Blood 2012, 119, 521–529. [Google Scholar] [CrossRef]
  14. Rossi, D.; Rasi, S.; Spina, V.; Fangazio, M.; Monti, S.; Greco, M.; Ciardullo, C.; Famà, R.; Cresta, S.; Bruscaggin, A.; et al. Different impact of NOTCH1 and SF3B1 mutations on the risk of chronic lymphocytic leukemia transformation to Richter syndrome. Br. J. Haematol. 2012, 158, 426–429. [Google Scholar] [CrossRef] [PubMed]
  15. Rossi, D.; Spina, V.; Cerri, M.; Rasi, S.; Deambrogi, C.; De Paoli, L.; Laurenti, L.; Maffei, R.; Forconi, F.; Bertoni, F.; et al. Stereotyped B-cell receptor is an independent risk factor of chronic lymphocytic leukemia transformation to Richter syndrome. Clin. Cancer Res. 2009, 15, 4415–4422. [Google Scholar] [CrossRef] [PubMed]
  16. Rossi, D.; Spina, V.; Deambrogi, C.; Rasi, S.; Laurenti, L.; Stamatopoulos, K.; Arcaini, L.; Lucioni, M.; Rocque, G.B.; Xu-Monette, Z.Y.; et al. The genetics of Richter syndrome reveals disease heterogeneity and predicts survival after transformation. Blood 2011, 117, 3391–3401. [Google Scholar] [CrossRef]
  17. Moreno, C.; Montserrat, E. New prognostic markers in chronic lymphocytic leukemia. Blood Rev. 2008, 22, 211–219. [Google Scholar] [CrossRef] [PubMed]
  18. Baliakas, P.; Jeromin, S.; Iskas, M.; Puiggros, A.; Plevova, K.; Nguyen-Khac, F.; Davis, Z.; Rigolin, G.M.; Visentin, A.; Xochelli, A.; et al. Cytogenetic complexity in chronic lymphocytic leukemia: Definitions, associations, and clinical impact. Blood 2019, 133, 1205–1216. [Google Scholar] [CrossRef]
  19. Ben-Dali, Y.; Hleuhel, M.H.; da Cunha-Bang, C.; Brieghel, C.; Poulsen, C.B.; Clasen-Linde, E.; Bentzen, H.H.N.; Frederiksen, H.; Christiansen, I.; Nielsen, L.H.; et al. Richter’s transformation in patients with chronic lymphocytic leukaemia: A Nationwide Epidemiological Study. Leuk. Lymphoma 2020, 61, 1435–1444. [Google Scholar] [CrossRef]
  20. Mao, Z.; Quintanilla-Martinez, L.; Raffeld, M.; Richter, M.; Krugmann, J.; Burek, C.; Hartmann, E.; Rudiger, T.; Jaffe, E.S.; Müller-Hermelink, H.K.; et al. IgVH Mutational Status and Clonality Analysis of Richter’s Transformation: Diffuse Large B-cell Lymphoma and Hodgkin Lymphoma in Association with B-cell Chronic Lymphocytic Leukemia (B-CLL) Represent 2 Different Pathways of Disease Evolution. Am. J. Surg. Pathol. 2007, 31, 1605–1614. [Google Scholar] [CrossRef]
  21. Parry, E.M.; ten Hacken, E.; Wu, C.J. Richter syndrome: Novel insights into the biology of transformation. Blood 2023, 142, 11–22. [Google Scholar] [CrossRef]
  22. Pepe, S.; Vitale, C.; Giannarelli, D.; Visentin, A.; Sanna, A.; Frustaci, A.M.; Olivieri, J.; Quaglia, F.M.; Gozzetti, A.; Sportoletti, P.; et al. Richter transformation in diffuse large B-cell lymphoma in patients with chronic lymphocytic leukemia receiving ibrutinib: Risk factors and outcomes. Leukemia 2025, 39, 1883–1891. [Google Scholar] [CrossRef] [PubMed]
  23. Nadeu, F.; Royo, R.; Massoni-Badosa, R.; Playa-Albinyana, H.; Garcia-Torre, B.; Duran-Ferrer, M.; Dawson, K.J.; Kulis, M.; Diaz-Navarro, A.; Villamor, N.; et al. Detection of early seeding of Richter transformation in chronic lymphocytic leukemia. Nat. Med. 2022, 28, 1662–1671. [Google Scholar] [CrossRef] [PubMed]
  24. Wang, X.; Chen, J. Molecular Features Accompanying Richter’s Transformation in Patients with Chronic Lymphocytic Leukemia. Int. J. Mol. Sci. 2025, 26, 5563. [Google Scholar] [CrossRef]
  25. Chakraborty, S.; Martines, C.; Porro, F.; Fortunati, I.; Bonato, A.; Dimishkovska, M.; Piazza, S.; Yadav, B.S.; Innocenti, I.; Fazio, R.; et al. B-cell receptor signaling and genetic lesions in TP53 and CDKN2A/CDKN2B cooperate in Richter transformation. Blood 2021, 138, 1053–1066. [Google Scholar] [CrossRef] [PubMed]
  26. Petrackova, A.; Turcsanyi, P.; Papajik, T.; Kriegova, E. Revisiting Richter transformation in the era of novel CLL agents. Blood Rev. 2021, 49, 100824. [Google Scholar] [CrossRef]
  27. Tsimberidou, A.M.; O’Brien, S.; Khouri, I.; Giles, F.J.; Kantarjian, H.M.; Champlin, R.; Wen, S.; Do, K.A.; Smith, S.C.; Lerner, S.; et al. Clinical outcomes and prognostic factors in patients with Richter’s syndrome treated with chemotherapy or chemoimmunotherapy with or without stem-cell transplantation. J. Clin. Oncol. 2006, 24, 2343–2351. [Google Scholar] [CrossRef]
  28. Al-Sawaf, O.; Robrecht, S.; Bahlo, J.; Fink, A.M.; Cramer, P.; v Tresckow, J.; Lange, E.; Kiehl, M.; Dreyling, M.; Ritgen, M.; et al. Richter transformation in chronic lymphocytic leukemia (CLL)-a pooled analysis of German CLL Study Group (GCLLSG) front line treatment trials. Leukemia 2021, 35, 169–176. [Google Scholar] [CrossRef]
  29. Elnair, R.; Ellithi, M.; Kallam, A.; Shostrom, V.; Bociek, R.G. Outcomes of Richter’s transformation of chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL): An analysis of the SEER database. Ann. Hematol. 2021, 100, 2513–2519. [Google Scholar] [CrossRef]
  30. Munir, T.; Brown, J.R.; O’Brien, S.; Barrientos, J.C.; Barr, P.M.; Reddy, N.M.; Coutre, S.; Tam, C.S.; Mulligan, S.P.; Jaeger, U.; et al. Final analysis from RESONATE: Up to six years of follow-up on ibrutinib in patients with previously treated chronic lymphocytic leukemia or small lymphocytic lymphoma. Am. J. Hematol. 2019, 94, 1353–1363. [Google Scholar] [CrossRef]
  31. Burger, J.A.; Barr, P.M.; Robak, T.; Owen, C.; Ghia, P.; Tedeschi, A.; Bairey, O.; Hillmen, P.; Coutre, S.E.; Devereux, S.; et al. Long-term efficacy and safety of first-line ibrutinib treatment for patients with CLL/SLL: 5 years of follow-up from the phase 3 RESONATE-2 study. Leukemia 2020, 34, 787–798. [Google Scholar] [CrossRef]
  32. Seymour, J.F.; Kipps, T.J.; Eichhorst, B.; Hillmen, P.; D’Rozario, J.; Assouline, S.; Owen, C.; Gerecitano, J.; Robak, T.; De la Serna, J.; et al. Venetoclax-Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2018, 378, 1107–1120. [Google Scholar] [CrossRef]
  33. Maddocks, K.J.; Ruppert, A.S.; Lozanski, G.; Heerema, N.A.; Zhao, W.; Abruzzo, L.; Lozanski, A.; Davis, M.; Gordon, A.; Smith, L.L.; et al. Etiology of Ibrutinib Therapy Discontinuation and Outcomes in Patients with Chronic Lymphocytic Leukemia. JAMA Oncol. 2015, 1, 80–87. [Google Scholar] [CrossRef]
  34. Jain, P.; Keating, M.; Wierda, W.; Estrov, Z.; Ferrajoli, A.; Jain, N.; George, B.; James, D.; Kantarjian, H.; Burger, J.; et al. Outcomes of patients with chronic lymphocytic leukemia after discontinuing ibrutinib. Blood 2015, 125, 2062–2067. [Google Scholar] [CrossRef]
  35. Davids, M.S.; Huang, Y.; Rogers, K.A.; Stern, R.; Brown, J.R.; Thompson, P.A.; Brander, D.M.; Danilov, A.V.; Ujjani, C.S.; Parikh, S.A.; et al. Richter’s syndrome (RS) in patients with chronic lymphocytic leukemia (CLL) on novel agent therapy. J. Clin. Oncol. 2017, 35 (Suppl. S15), 7505. [Google Scholar] [CrossRef]
  36. Appleby, N.; Eyre, T.A.; Cabes, M.; Jackson, A.; Boucher, R.; Yates, F.; Fox, S.; Rawstron, A.; Hillmen, P.; Schuh, A. The STELLAR trial protocol: A prospective multicentre trial for Richter’s syndrome consisting of a randomised trial investigation CHOP-R with or without acalabrutinib for newly diagnosed RS and a single-arm platform study for evaluation of novel agents in relapsed disease. BMC Cancer 2019, 19, 471. [Google Scholar]
  37. Rawstron, A.C.; Fazi, C.; Agathangelidis, A.; Villamor, N.; Letestu, R.; Nomdedeu, J.; Palacio, C.; Stehlikova, O.; Kreuzer, K.A.; Liptrot, S.; et al. A complementary role of multiparameter flow cytometry and high-throughput sequencing for minimal residual disease detection in chronic lymphocytic leukemia: An European Research Initiative on CLL study. Leukemia 2016, 30, 929–936. [Google Scholar] [CrossRef] [PubMed]
  38. Thompson, P.A.; Srivastava, J.; Peterson, C.; Strati, P.; Jorgensen, J.L.; Hether, T.; Keating, M.J.; O’Brien, S.M.; Ferrajoli, A.; Burger, J.A.; et al. Minimal residual disease undetectable by next-generation sequencing predicts improved outcome in CLL after chemoimmunotherapy. Blood 2019, 134, 1951–1959. [Google Scholar] [CrossRef]
  39. Feng, S.; Zheng, P.; Liu, H.; Sun, M.; Guo, Y.; Ma, L.; Liu, R.; Fu, Z.; Yang, F.; Ke, X.; et al. Chimeric antigen receptor (CAR)-T cell therapy treatment Richter’s transformation. Front. Oncol. 2025, 15, 1591360. [Google Scholar] [CrossRef] [PubMed]
  40. Coombs, C.C.; Easaw, S.; Grover, N.S.; O’Brien, S.M. Cellular Therapies in Chronic Lymphocytic Leukemia and Richter’s Transformation: Recent Developments in Chimeric Antigen Receptor T-Cells, Natural Killer Cells, and Allogeneic Stem Cell Transplant. Cancers 2023, 15, 1838. [Google Scholar] [CrossRef] [PubMed]
  41. Guièze, R.; Eikema, D.J.; Koster, L.; Schetelig, J.; Sengeloev, H.; Passweg, J.; Finke, J.; Arat, M.; Broers, A.E.C.; Stölzel, F.; et al. Allogeneic hematopoietic stem-cell transplantation for patients with Richter transformation: A retrospective study on behalf of the Chronic Malignancies Working Party of the EBMT. Bone Marrow Transplant. 2024, 59, 950–956. [Google Scholar] [CrossRef]
  42. Ryan, C.E.; Davids, M.S. Practical Management of Richter Transformation in 2023 and Beyond. Am. Soc. Clin. Oncol. Educ. Book 2023, 43, e390804. [Google Scholar] [CrossRef] [PubMed]
  43. Langerbeins, P.; Busch, R.; Anheier, N.; Dürig, J.; Bergmann, M.; Goebeler, M.E.; Hurtz, H.J.; Stauch, M.B.; Stilgenbauer, S.; Döhner, H.; et al. Poor efficacy and tolerability of R-CHOP in relapsed/refractory chronic lymphocytic leukemia and Richter transformation. Am. J. Hematol. 2014, 89, E239–E243. [Google Scholar] [CrossRef]
  44. Durot, E.; Michallet, A.-S.; Leprêtre, S.; Le, Q.-H.; Leblond, V.; Delmer, A. Platinum and high-dose cytarabine-based regimens are efficient in ultra high/high-risk chronic lymphocytic leukemia and Richter’s syndrome: Results of a French retrospective multicenter study. Eur. J. Haematol. 2015, 95, 160–167. [Google Scholar] [CrossRef] [PubMed]
  45. Dabaja, B.S.; O’Brien, S.M.; Kantarjian, H.M.; Cortes, J.E.; Thomas, D.A.; Albitar, M.; Schlette, E.S.; Faderl, S.; Sarris, A.; Keating, M.J.; et al. Fractionated cyclophosphamide, vincristine, liposomal daunorubicin (daunoXome), and dexamethasone (hyperCVXD) regimen in Richter’s syndrome. Leuk. Lymphoma 2001, 42, 329–337. [Google Scholar] [CrossRef] [PubMed]
  46. Tsimberidou, A.M.; Kantarjian, H.M.; Cortes, J.; Thomas, D.A.; Faderl, S.; Garcia-Manero, G.; Verstovsek, S.; Ferrajoli, A.; Wierda, W.; Alvarado, Y.; et al. Fractionated cyclophosphamide, vincristine, liposomal daunorubicin, and dexamethasone plus rituximab and granulocyte-macrophage-colony stimulating factor (GM-CSF) alternating with methotrexate and cytarabine plus rituximab and GM-CSF in patients with Richter syndrome or fludarabine-refractory chronic lymphocytic leukemia. Cancer 2003, 97, 1711–1720. [Google Scholar]
  47. Tsimberidou, A.M.; Wierda, W.G.; Plunkett, W.; Kurzrock, R.; O’Brien, S.; Wen, S.; Ferrajoli, A.; Ravandi-Kashani, F.; Garcia-Manero, G.; Estrov, Z.; et al. Phase I-II study of oxaliplatin, fludarabine, cytarabine, and rituximab combination therapy in patients with Richter’s syndrome or fludarabine-refractory chronic lymphocytic leukemia. J. Clin. Oncol. 2008, 26, 196–203. [Google Scholar] [CrossRef] [PubMed]
  48. Rogers, K.A.; Huang, Y.; Ruppert, A.S.; Salem, G.; Stephens, D.M.; Heerema, N.A.; Andritsos, L.A.; Awan, F.T.; Byrd, J.C.; Flynn, J.M.; et al. A single-institution retrospective cohort study of first-line R-EPOCH chemoimmunotherapy for Richter syndrome demonstrating complex chronic lymphocytic leukaemia karyotype as an adverse prognostic factor. Br. J. Haematol. 2018, 180, 259–266. [Google Scholar] [CrossRef]
  49. Tsang, M.; Shanafelt, T.D.; Call, T.G.; Ding, W.; Chanan-Khan, A.; Leis, J.F.; Nowakowski, G.S.; Bowen, D.; Conte, M.; Schwager, S.M.; et al. The efficacy of ibrutinib in the treatment of Richter syndrome. Blood 2015, 125, 1676–1678. [Google Scholar] [CrossRef]
  50. Maschalidi, S.; Sepulveda, F.E.; Garrigue, A.; Fischer, A.; de Saint Basile, G. Therapeutic effect of JAK1/2 blockade on the manifestations of hemophagocytic lymphohistiocytosis in mice. Blood 2016, 128, 60–71. [Google Scholar] [CrossRef]
  51. Eyre, T.A.; Schuh, A.; Wierda, W.G.; Brown, J.R.; Ghia, P.; Pagel, J.M.; Furman, R.R.; Cheung, J.; Hamdy, A.; Izumi, R.; et al. Acalabrutinib monotherapy for treatment of chronic lymphocytic leukaemia (ACE-CL-001): Analysis of the Richter transformation cohort of an open-label, single-arm, phase 1-2 study. Lancet Haematol. 2021, 8, e912–e921. [Google Scholar] [CrossRef]
  52. Al-Sawaf, O.; Ligtvoet, R.; Robrecht, S.; Stumpf, J.; Fink, A.M.; Tausch, E.; Schneider, C.; Boettcher, S.; Mikusko, M.; Ritgen, M.; et al. Tislelizumab plus zanubrutinib for Richter transformation: The phase 2 RT1 trial. Nat. Med. 2024, 30, 240–248. [Google Scholar] [CrossRef]
  53. Wierda, W.G.; Shah, N.N.; Cheah, C.Y.; Lewis, D.; Hoffmann, M.S.; Coombs, C.C.; Lamanna, N.; Ma, S.; Jagadeesh, D.; Munir, T.; et al. Pirtobrutinib, a highly selective, non-covalent (reversible) BTK inhibitor in patients with B-cell malignancies: Analysis of the Richter transformation subgroup from the multicentre, open-label, phase 1/2 BRUIN study. Lancet Haematol. 2024, 11, e682–e692. [Google Scholar] [CrossRef]
  54. Del Gaizo Moore, V.; Brown, J.R.; Certo, M.; Love, T.M.; Novina, C.D.; Letai, A. Chronic lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737. J. Clin. Investig. 2007, 117, 112–121. [Google Scholar] [CrossRef]
  55. Davids, M.S.; Roberts, A.W.; Seymour, J.F.; Pagel, J.M.; Kahl, B.S.; Wierda, W.G.; Puvvada, S.; Kipps, T.J.; Anderson, M.A.; Salem, A.H.; et al. Phase I First-in-Human Study of Venetoclax in Patients with Relapsed or Refractory Non-Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 826–833. [Google Scholar] [CrossRef]
  56. Davids, M.S.; Rogers, K.A.; Tyekucheva, S.; Wang, Z.; Pazienza, S.; Renner, S.K.; Montegaard, J.; Ihuoma, U.; Lehmberg, T.Z.; Parry, E.M.; et al. Venetoclax plus dose-adjusted R-EPOCH for Richter syndrome. Blood 2022, 139, 686–689. [Google Scholar] [CrossRef]
  57. Davids, M.S.; Rogers, K.A.; Jain, N.; Tyekucheva, S.; Ren, Y.; Carey, C.; Montegaard, J.; Hajdenberg, M.; Ryan, C.E.; Merryman, R.; et al. Initial results of a multicenter phase 2 study of venetoclax in combination with R-CHOP (VR-CHOP) for patients with Richter Syndrome. Hematol. Oncol. 2023, 41 (Suppl. S2), 466–468. [Google Scholar] [CrossRef]
  58. Hampel, P.J.; Swaminathan, M.; Rogers, K.A.; Parry, E.M.; Burger, J.A.; Davids, M.S.; Ding, W.; Ferrajoli, A.; Hyak, J.M.; Jain, N.; et al. A multicenter study of venetoclax-based treatment for patients with Richter transformation of chronic lymphocytic leukemia. Blood Adv. 2024, 8, 2342–2350. [Google Scholar] [CrossRef]
  59. He, R.; Ding, W.; Viswanatha, D.S.; Chen, D.; Shi, M.; Van Dyke, D.; Tian, S.; Dao, L.N.; Parikh, S.A.; Shanafelt, T.D.; et al. PD-1 Expression in Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (CLL/SLL) and Large B-cell Richter Transformation (DLBCL-RT): A Characteristic Feature of DLBCL-RT and Potential Surrogate Marker for Clonal Relatedness. Am. J. Surg. Pathol. 2018, 42, 843–854. [Google Scholar] [CrossRef] [PubMed]
  60. Frustaci, A.M.; Montillo, M.; Rossi, D.; Zinzani, P.L.; Motta, M.; Gaidano, G.; Quaresmini, G.; Scarfo, L.; Pietrasanta, D.; Coscia, M.; et al. Results of MOLTO, a multicenter, open label, phase II clinical trial evaluating venetoclax, atezolizumab and obinutuzumab combination in Richter syndrome. J. Clin. Oncol. 2023, 41 (Suppl. S16), 7502. [Google Scholar] [CrossRef]
  61. Iannello, A.; Vitale, N.; Coma, S.; Arruga, F.; Chadburn, A.; Di Napoli, A.; Laudanna, C.; Allan, J.N.; Furman, R.R.; Pachter, J.A.; et al. Synergistic efficacy of the dual PI3K-δ/γ inhibitor duvelisib with the Bcl-2 inhibitor venetoclax in Richter syndrome PDX models. Blood 2021, 137, 3378–3389. [Google Scholar] [CrossRef] [PubMed]
  62. Crombie, J.L.; Ryan, C.E.; Ren, Y.; Tyekucheva, S.; Carey, C.; Zou, A.; Normilus, S.; Montegaard, J.; Soumerai, J.D.; Bhandari, S.; et al. A Phase 2 Study of Duvelisib and Venetoclax in Patients with Relapsed or Refractory (R/R) Chronic Lymphocytic Leukemia (CLL) or Richter’s Syndrome (RS). Blood 2024, 144 (Suppl. S1), 4628. [Google Scholar] [CrossRef]
  63. Siddiqi, T. Chronic Lymphocytic Leukemia (CLL): Biology and Therapy. In Biology and Treatment of Leukemia and Bone Marrow Neoplasms; Pullarkat, V., Marcucci, G., Eds.; Springer International Publishing: Cham, Switzerland, 2021; pp. 133–149. [Google Scholar]
  64. Kharfan-Dabaja, M.A.; Kumar, A.; Stingo, F.E.; Khimani, F.; Hussaini, M.; Ayala, E.; Nishihori, T.; Shah, B.; Locke, F.L.; Pinilla-Ibarz, J.; et al. Allogeneic Hematopoietic Cell Transplantation for Richter Syndrome: A Single-Center Experience. Clin. Lymphoma Myeloma Leuk. 2018, 18, e35–e39. [Google Scholar] [CrossRef]
  65. Sośnia, O.; Pruszczyk, K.; Danecki, M.; Wąsik-Szczepanek, E.; Tryc-Szponder, J.; Iskierka-Jażdżewska, E.; Majeranowski, A.; Krzemień, H.; Bołkun, Ł.; Paszkiewicz-Kozik, E.; et al. Richter transformation—Retrospective treatment outcomes analysis in Polish Adult Leukemia Study Group. Leuk. Lymphoma 2024, 65, 175–186. [Google Scholar] [CrossRef] [PubMed]
  66. Lahoud, O.B.; Devlin, S.M.; Maloy, M.A.; Roeker, L.E.; Dahi, P.B.; Ponce, D.M.; Gyurkocza, B.; Koehne, G.; Young, J.W.; Castro-Malaspina, H.R.; et al. Reduced-intensity conditioning hematopoietic stem cell transplantation for chronic lymphocytic leukemia and Richter’s transformation. Blood Adv. 2021, 5, 2879–2889. [Google Scholar] [CrossRef]
  67. Backhaus, D.; Brauer, D.; Pointner, R.; Bischof, L.; Vucinic, V.; Franke, G.-N.; Niederwieser, D.; Platzbecker, U.; Jentzsch, M.; Schwind, S. A high hematopoietic cell transplantation comorbidity Index (HCT-CI) does not impair outcomes after non-myeloablative allogeneic stem cell transplantation in acute myeloid leukemia patients 60 years or older. Bone Marrow Transplant. 2023, 58, 30–38. [Google Scholar] [CrossRef]
  68. Janscak, M.; Stelmes, A.; van den Berg, J.; Heim, D.; Halter, J.; Drexler, B.; Arranto, C.; Passweg, J.; Medinger, M. Influence of comorbidities on outcome in 1102 patients with an allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2024, 59, 1525–1533. [Google Scholar] [CrossRef]
  69. Gao, L.; Yang, L.; Zhou, S.; Zhu, W.; Han, Y.; Chen, S.; Xue, S.; Wang, Y.; Qiu, H.; Wu, D.; et al. Allogenic hematopoietic stem cell transplantation outcomes of patients aged ≥55 years with acute myeloid leukemia or myelodysplastic syndromes in China: A retrospective study. Stem Cell Res. Ther. 2024, 15, 24. [Google Scholar] [CrossRef]
  70. Thakar, M.S.; Broglie, L.; Logan, B.; Artz, A.; Bunin, N.; Burroughs, L.M.; Fretham, C.; Jacobsohn, D.A.; Loren, A.W.; Kurtzberg, J.; et al. The Hematopoietic Cell Transplant Comorbidity Index predicts survival after allogeneic transplant for nonmalignant diseases. Blood 2019, 133, 754–762. [Google Scholar] [CrossRef] [PubMed]
  71. Herrera, A.F.; Ahn, K.W.; Litovich, C.; Chen, Y.; Assal, A.; Bashir, Q.; Bayer, R.L.; Coleman, M.; DeFilipp, Z.; Farhadfar, N.; et al. Autologous and allogeneic hematopoietic cell transplantation for diffuse large B-cell lymphoma-type Richter syndrome. Blood Adv. 2021, 5, 3528–3539. [Google Scholar] [CrossRef]
  72. Puckrin, R.; Owen, C.; Fontaine, A.; Peters, A.; Stewart, D.; Shafey, M. Allogeneic hematopoietic cell transplantation for Richter transformation of chronic lymphocytic leukemia: An intention-to-transplant analysis. Bone Marrow Transpl. 2023, 58, 817–819. [Google Scholar] [CrossRef]
  73. Cwynarski, K.; van Biezen, A.; de Wreede, L.; Stilgenbauer, S.; Bunjes, D.; Metzner, B.; Koza, V.; Mohty, M.; Remes, K.; Russell, N.; et al. Autologous and allogeneic stem-cell transplantation for transformed chronic lymphocytic leukemia (Richter’s syndrome): A retrospective analysis from the chronic lymphocytic leukemia subcommittee of the chronic leukemia working party and lymphoma working party of the European Group for Blood and Marrow Transplantation. J. Clin. Oncol. 2012, 30, 2211–2217. [Google Scholar] [PubMed]
  74. El-Asmar, J.; Kharfan-Dabaja, M.A. Hematopoietic Cell Transplantation for Richter Syndrome. Biol. Blood Marrow Transplant. 2016, 22, 1938–1944. [Google Scholar] [CrossRef]
  75. Bouclet, F.; Calleja, A.; Dilhuydy, M.S.; Véronèse, L.; Pereira, B.; Amorim, S.; Cymbalista, F.; Herbaux, C.; de Guibert, S.; Roos-Weil, D.; et al. Real-world outcomes following venetoclax therapy in patients with chronic lymphocytic leukemia or Richter syndrome: A FILO study of the French compassionate use cohort. Ann. Hematol. 2021, 100, 987–993. [Google Scholar] [CrossRef]
  76. Español, I.; Büchler, T.; Ferrá, C.; Gallardo, D.; Reyes, P.; Sarrá, J.; Domingo, A.; Romagosa, V.; Grañena, A. Richter’s syndrome after allogeneic stem cell transplantation for chronic lymphocytic leukaemia successfully treated by withdrawal of immunosuppression, and donor lymphocyte infusion. Bone Marrow Transpl. 2003, 31, 215–218. [Google Scholar] [CrossRef][Green Version]
  77. Kim, H.T.; Baker, P.O.; Parry, E.; Davids, M.; Alyea, E.P.; Ho, V.T.; Cutler, C.; Koreth, J.; Gooptu, M.; Romee, R.; et al. Allogeneic hematopoietic cell transplantation outcomes in patients with Richter’s transformation. Haematologica 2021, 106, 3219–3222. [Google Scholar] [CrossRef]
  78. Beer, S.A.; Blättel, J.; Reuß, K.; Maier, C.-P.; Faul, C.; Vogel, W.; Bethge, W.; Lengerke, C. Long-term patient-reported outcomes following allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2025, 60, 617–624. [Google Scholar] [CrossRef]
  79. Neelapu, S.S.; Locke, F.L.; Bartlett, N.L.; Lekakis, L.J.; Miklos, D.B.; Jacobson, C.A.; Braunschweig, I.; Oluwole, O.O.; Siddiqi, T.; Lin, Y.; et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N. Engl. J. Med. 2017, 377, 2531–2544. [Google Scholar] [CrossRef] [PubMed]
  80. Schuster, S.J.; Bishop, M.R.; Tam, C.S.; Waller, E.K.; Borchmann, P.; McGuirk, J.P.; Jäger, U.; Jaglowski, S.; Andreadis, C.; Westin, J.R.; et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2019, 380, 45–56. [Google Scholar] [CrossRef] [PubMed]
  81. Benjamini, O.; Fried, S.; Shouval, R.; Flynn, J.R.; Beyar-Katz, O.; Leslie, L.A.; Zucherman, T.; Yerushalmi, R.; Shem-Tov, N.; Palomba, M.L.; et al. Anti-CD19 chimeric antigen receptor T-cell therapy has less efficacy in Richter transformation than in de novo large B-cell lymphoma and transformed low-grade B-cell lymphoma. Haematologica 2024, 109, 3566–3577. [Google Scholar] [CrossRef]
  82. Kittai, A.S.; Bond, D.; Huang, Y.; Bhat, S.A.; Blyth, E.; Byrd, J.C.; Chavez, J.C.; Davids, M.S.; Cruz, J.P.D.; Dowling, M.R.; et al. Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy for Richter Transformation: An International, Multicenter, Retrospective Study. J. Clin. Oncol. 2024, 42, 2071–2079. [Google Scholar] [CrossRef]
  83. Beyar-Katz, O.; Benjamini, O.; Delgado, J.; Ruella, M.; Ram, R.; Grisariu, S.; Visentin, A.; Vandenberghe, E.; Gentile, M.; Avigdor, A.; et al. CD19 CAR-T Cell Therapy Is Effective in Richter Transformation: A Multicenter Retrospective Analysis By the European Research Initiative on Chronic Lymphocytic Leukemia. Blood 2024, 144 (Suppl. S1), 4504. [Google Scholar] [CrossRef]
  84. Kittai, A.S.; Bond, D.A.; William, B.; Saad, A.; Penza, S.; Efebera, Y.; Larkin, K.; Wall, S.A.; Choe, H.K.; Bhatnagar, B.; et al. Clinical activity of axicabtagene ciloleucel in adult patients with Richter syndrome. Blood Adv. 2020, 4, 4648–4652. [Google Scholar] [CrossRef] [PubMed]
  85. Abramson, J.S.; Palomba, M.L.; Gordon, L.I.; Lunning, M.A.; Wang, M.; Arnason, J.; Mehta, A.; Purev, E.; Maloney, D.G.; Andreadis, C.; et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): A multicentre seamless design study. Lancet 2020, 396, 839–852. [Google Scholar] [CrossRef]
  86. European Medicines Agency. Breyanzi (Lisocabtagene Maraleucel)—EPAR Product Information. 2022. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/breyanzi (accessed on 20 July 2025).
  87. Siddiqi, T.; Maloney, D.G.; Kenderian, S.S.; Brander, D.M.; Dorritie, K.; Soumerai, J.; Riedell, P.A.; Shah, N.N.; Nath, R.; Fakhri, B.; et al. Lisocabtagene maraleucel in chronic lymphocytic leukaemia and small lymphocytic lymphoma (TRANSCEND CLL 004): A multicentre, open-label, single-arm, phase 1–2 study. Lancet 2023, 402, 641–654. [Google Scholar] [CrossRef]
  88. Kamdar, M.; Solomon, S.R.; Arnason, J.; Johnston, P.B.; Glass, B.; Bachanova, V.; Ibrahimi, S.; Mielke, S.; Mutsaers, P.; Hernandez-Ilizaliturri, F.; et al. Lisocabtagene maraleucel versus standard of care with salvage chemotherapy followed by autologous stem cell transplantation as second-line treatment in patients with relapsed or refractory large B-cell lymphoma (TRANSFORM): Results from an interim analysis of an open-label, randomised, phase 3 trial. Lancet 2022, 399, 2294–2308, Erratum in Lancet 2022, 400, 160. [Google Scholar]
  89. Fraietta, J.A.; Lacey, S.F.; Orlando, E.J.; Pruteanu-Malinici, I.; Gohil, M.; Lundh, S.; Boesteanu, A.C.; Wang, Y.; O’Connor, R.S.; Hwang, W.T.; et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat. Med. 2021, 27, 561, Erratum in Nat. Med. 2018, 24, 563–571. [Google Scholar] [CrossRef] [PubMed]
  90. Jain, N.; Senapati, J.; Thakral, B.; Ferrajoli, A.; Thompson, P.; Burger, J.; Basu, S.; Kadia, T.; Daver, N.; Borthakur, G.; et al. A phase 2 study of nivolumab combined with ibrutinib in patients with diffuse large B-cell Richter transformation of CLL. Blood Adv. 2023, 7, 1958–1966. [Google Scholar] [CrossRef]
  91. Ding, W.; LaPlant, B.R.; Call, T.G.; Parikh, S.A.; Leis, J.F.; He, R.; Shanafelt, T.D.; Sinha, S.; Le-Rademacher, J.; Feldman, A.L.; et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 2017, 129, 3419–3427. [Google Scholar] [CrossRef]
  92. Younes, A.; Brody, J.; Carpio, C.; Lopez-Guillermo, A.; Ben-Yehuda, D.; Ferhanoglu, B.; Nagler, A.; Ozcan, M.; Avivi, I.; Bosch, F.; et al. Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non-Hodgkin lymphoma or chronic lymphocytic leukaemia: A phase 1/2a study. Lancet Haematol. 2019, 6, e67–e78. [Google Scholar] [CrossRef] [PubMed]
  93. Nadiminti, K.V.; Ahn, K.W.; Patel, D.J.; Lian, Q.; Pasquini, M.C.; Hamadani, M.; Turtle, C.J.; Herrera, A.F.; Shadman, M. CD19-Directed CAR-T Therapy for Richter Transformation: A CIBMTR Analysis. Transplant. Cell. Ther. 2025, 31 (Suppl. S2), S207–S208. [Google Scholar] [CrossRef]
  94. Barata, A.; Hoogland, A.I.; Kommalapati, A.; Logue, J.; Welniak, T.; Hyland, K.A.; Eisel, S.L.; Small, B.J.; Jayani, R.V.; Booth-Jones, M.; et al. Change in Patients’ Perceived Cognition Following Chimeric Antigen Receptor T-Cell Therapy for Lymphoma. Transplant. Cell Ther. 2022, 28, e1–e401. [Google Scholar] [CrossRef]
  95. Ruark, J.; Mullane, E.; Cleary, N.; Cordeiro, A.; Bezerra, E.D.; Wu, V.; Voutsinas, J.; Shaw, B.E.; Flynn, K.E.; Lee, S.J.; et al. Patient-Reported Neuropsychiatric Outcomes of Long-Term Survivors after Chimeric Antigen Receptor T Cell Therapy. Biol. Blood Marrow Transplant. 2020, 26, 34–43. [Google Scholar] [CrossRef]
  96. Amonoo, H.L.; Johnson, P.C.; Dhawale, T.M.; Traeger, L.; Rice, J.; Lavoie, M.W.; Ufere, N.N.; Longley, R.M.; Harnedy, L.E.; Clay, M.A.; et al. Sharing and caring: The impact of social support on quality of life and health outcomes in hematopoietic stem cell transplantation. Cancer 2021, 127, 1260–1265. [Google Scholar] [CrossRef] [PubMed]
  97. Dai, H.; Xu, S.; Wang, Z.; Li, Z.; Cao, J.; Hu, T.; Zhou, F. Quality of life and symptom burden among hematologic malignancy patients undergoing CAR-T therapy: A cross-sectional study. Sci. Rep. 2025, 15, 17763. [Google Scholar] [CrossRef] [PubMed]
  98. Abramson, J.S.; Palomba, M.L.; Gordon, L.I.; Lunning, M.; Wang, M.; Arnason, J.; Purev, E.; Maloney, D.G.; Andreadis, C.; Sehgal, A.; et al. Two-year follow-up of lisocabtagene maraleucel in relapsed or refractory large B-cell lymphoma in TRANSCEND NHL 001. Blood 2024, 143, 404–416. [Google Scholar] [CrossRef]
  99. Locke, F.L.; Ghobadi, A.; Jacobson, C.A.; Miklos, D.B.; Lekakis, L.J.; Oluwole, O.O.; Lin, Y.; Braunschweig, I.; Hill, B.T.; Timmerman, J.M.; et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): A single-arm, multicentre, phase 1–2 trial. Lancet Oncol. 2019, 20, 31–42. [Google Scholar] [CrossRef] [PubMed]
  100. Vom Stein, A.F.; Nguyen, P.H.; Ten Hacken, E. A question of TiME: How microenvironmental interactions shape response to immunotherapy in CLL and Richter Transformation. Front. Immunol. 2025, 16, 1592574. [Google Scholar] [CrossRef]
  101. Levy Yurkovski, I.; Tadmor, T. Accelerated Chronic Lymphocytic Leukemia and Richter Transformation in the Era of Novel Agents. Acta Haematol. 2024, 147, 73–83. [Google Scholar] [CrossRef] [PubMed]
  102. Ortiz-Maldonado, V.; Martínez-Cibrián, N.; Alserawan, L.; Betriu, S.; Triguero, A.; Blum, S.; Faes, M.; Liefaard, M.C.; Pont, M.J.; Spoon, M.; et al. Euplagia-1: A Phase 1/2 Trial of GLPG5201, a Fresh Stem-like Early Memory CD19 CAR T-Cell Therapy with a 7-Day Vein-to-Vein Time, in Patients with Relapsed/Refractory CLL and RT. Blood 2024, 144 (Suppl. S1), 3452. [Google Scholar] [CrossRef]
  103. Kater, A.P.; Janssens, A.; Eradat, H.; Offner, F.; Sandoval-Sus, J.D.; Shadman, M.; Bjørn Poulsen, C.; Haaber Christensen, J.; Thompson, M.C.; Rios, M.; et al. CLL-280 Epcoritamab Induces Deep Responses in Patients with Richter Transformation (RT): Primary Results From the EPCORE CLL-1 Trial. Clin. Lymphoma Myeloma Leuk. 2024, 24, S350–S351. [Google Scholar] [CrossRef]
  104. Cheah, C.Y.; Assouline, S.; Baker, R.; Bartlett, N.L.; El-Sharkawi, D.; Giri, P.; Ku, M.; Schuster, S.J.; Matasar, M.; Radford, J.; et al. Mosunetuzumab Monotherapy Demonstrates Activity and a Manageable Safety Profile in Patients with Relapsed or Refractory Richter’s Transformation. Blood 2023, 142, 614. [Google Scholar] [CrossRef]
  105. Guièze, R.; Ysebaert, L.; Roos-Weil, D.; Fornecker, L.-M.; Ferrant, E.; Molina, L.; Aurran, T.; Clavert, A.; de Guibert, S.; Michallet, A.-S.; et al. Blinatumomab after R-CHOP bridging therapy for patients with Richter transformation: A phase 2 multicentre trial. Nat. Commun. 2024, 15, 6822. [Google Scholar] [CrossRef]
  106. Tedeschi, A.; Frustaci, A.M.; Condoluci, A.; Coscia, M.; Chiarle, R.; Zinzani, P.L.; Motta, M.; Gaidano, G.; Quaresmini, G.; Scarfò, L.; et al. Atezolizumab, venetoclax, and obinutuzumab combination in Richter transformation diffuse large B-cell lymphoma (MOLTO): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 2024, 25, 1298–1309. [Google Scholar] [CrossRef]
  107. Shouse, G.; Chen, C.; Muir, A.; Popplewell, L.; Siddiqi, T.; Zain, J.; Herrera, A.F.; Danilova, O.; Roleder, C.; Wang, L.; et al. Safety and efficacyof the combination of copanlisib and nivolumab in patients with Richter’s transformation or transformed non-Hodgkin lymphoma: Results from a phase I trial. Haematologica 2025. [Google Scholar] [CrossRef]
  108. Wang, Y.; LaPlant, B.R.; Parikh, S.A.; Leis, J.F.; Call, T.G.; Shanafelt, T.D.; He, R.; Micallef, I.N.; Nowakowski, G.S.; Habermann, T.M.; et al. Efficacy of pembrolizumab monotherapy and in combination with BCR inhibitors for Richter transformation of chronic lymphocytic leukemia (CLL). J. Clin. Oncol. 2024, 42 (Suppl. S16), 7050. [Google Scholar] [CrossRef]
  109. Huntington, S.F.; Schuster, S.J.; Ding, W.; Koehler, A.B.; Brander, D.M.; Rosenthal, A.C.; Leis, J.F.; Tun, H.W.; Moustafa, M.A.; Iqbal, M.; et al. DTRMWXHS-12, a novel Bruton tyrosine kinase inhibitor, in combination with everolimus and pomalidomide in patients with relapsed/refractory lymphomas: An open-label, multicenter, phase 1a/1b study. Am. J. Hematol. 2023, 98, 739–749. [Google Scholar] [CrossRef] [PubMed]
  110. A Study of DTRM-555 in Patients with Relapsed/Refractory Hematologic Malignancies. ClinicalTrials.gov, 2025. Available online: https://clinicaltrials.gov/study/NCT04305444?intr=DTRM-555&rank=1 (accessed on 20 July 2025).
  111. Stock, S.; Kluever, A.K.; Endres, S.; Kobold, S. Enhanced Chimeric Antigen Receptor T Cell Therapy through Co-Application of Synergistic Combination Partners. Biomedicines 2022, 10, 307. [Google Scholar] [CrossRef] [PubMed]
  112. Bouwstra, R.; van Meerten, T.; Bremer, E. CD47-SIRPα blocking-based immunotherapy: Current and prospective therapeutic strategies. Clin. Transl. Med. 2022, 12, e943. [Google Scholar] [CrossRef] [PubMed]
  113. Bajwa, A.; Habib, A.; Kittai, A.S. Treatment of Richter’s Transformation with Novel Therapies. Curr. Hematol. Malig. Rep. 2024, 19, 45–55. [Google Scholar] [CrossRef]
  114. Clubb, J.D.; Gao, T.A.; Chen, Y.Y. Synthetic Biology in the Engineering of CAR-T and CAR-NK Cell Therapies: Facts and Hopes. Clin. Cancer Res. 2023, 29, 1390–1402. [Google Scholar] [CrossRef] [PubMed]
  115. Lewis, R.I.; Vom Stein, A.F.; Hallek, M. Targeting the tumor microenvironment for treating double-refractory chronic lymphocytic leukemia. Blood 2024, 144, 601–614. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, M.; Mei, M.; Barr, P.M.; Barrientos, J.; de Vos, S.; Furman, R.; Patel, K.; Thompson, P.A.; Choi, M.; Kallam, A.; et al. Phase 1 Dose Escalation and Cohort Expansion Study of the Anti-ROR1 Antibody-Drug Conjugate Zilovertamab Vedotin (MK-2140) for the Treatment of Non-Hodgkin Lymphoma. Blood 2021, 138, 528. [Google Scholar] [CrossRef]
Figure 1. Molecular mechanisms underlying Richter transformation. This diagram illustrates how chronic B-cell receptor (BCR) activation interacts with recurrent genetic lesions to drive the pathogenesis of Richter transformation (RT). BCR signaling—often delivered through unmutated or stereotyped IGHV receptors (e.g., subset #8)—activates downstream pathways such as PI3K–AKT and NF-κB. Loss of tumor-suppressor checkpoints (TP53 mutation/deletion and CDKN2A/B loss) removes cell-cycle brakes and apoptotic safeguards, enabling BCR-dependent proliferation of large pleomorphic cells. NOTCH1 mutations, which are associated with biased subset-8 BCR usage and autonomous signaling, potentiate PI3K/AKT and NF-κB pathway activation and markedly increase the risk of transformation. These pathways converge on MYC, promoting metabolic reprogramming and rapid proliferation. Constitutive AKT phosphorylation—observed in high-risk CLL with NOTCH1 or TP53 alterations—further underscores the interplay between BCR, PI3K/AKT and NOTCH1 signaling. Together, chronic antigenic drive and cumulative genetic lesions produce profound genomic instability (chromothripsis, chromoplexy, and whole-genome doubling), resulting in the emergence of aggressive RT clones.
Figure 1. Molecular mechanisms underlying Richter transformation. This diagram illustrates how chronic B-cell receptor (BCR) activation interacts with recurrent genetic lesions to drive the pathogenesis of Richter transformation (RT). BCR signaling—often delivered through unmutated or stereotyped IGHV receptors (e.g., subset #8)—activates downstream pathways such as PI3K–AKT and NF-κB. Loss of tumor-suppressor checkpoints (TP53 mutation/deletion and CDKN2A/B loss) removes cell-cycle brakes and apoptotic safeguards, enabling BCR-dependent proliferation of large pleomorphic cells. NOTCH1 mutations, which are associated with biased subset-8 BCR usage and autonomous signaling, potentiate PI3K/AKT and NF-κB pathway activation and markedly increase the risk of transformation. These pathways converge on MYC, promoting metabolic reprogramming and rapid proliferation. Constitutive AKT phosphorylation—observed in high-risk CLL with NOTCH1 or TP53 alterations—further underscores the interplay between BCR, PI3K/AKT and NOTCH1 signaling. Together, chronic antigenic drive and cumulative genetic lesions produce profound genomic instability (chromothripsis, chromoplexy, and whole-genome doubling), resulting in the emergence of aggressive RT clones.
Ijms 26 08747 g001
Figure 2. Targeted therapies under investigation for Richter transformation. The diagram highlights key therapeutic targets on transformed B cells and associated immune or signaling interactions. Shown are CD19-directed CAR T-cell products, CD20×CD3 bispecific antibodies, checkpoint inhibitors (PD-1/PD-L1), CD47 blockade, antibody–drug conjugates (ADCs), and small-molecule inhibitors targeting BCL2, BTK, PI3K, and mTOR. These agents represent mechanistically distinct approaches being combined to improve outcomes in Richter transformation.
Figure 2. Targeted therapies under investigation for Richter transformation. The diagram highlights key therapeutic targets on transformed B cells and associated immune or signaling interactions. Shown are CD19-directed CAR T-cell products, CD20×CD3 bispecific antibodies, checkpoint inhibitors (PD-1/PD-L1), CD47 blockade, antibody–drug conjugates (ADCs), and small-molecule inhibitors targeting BCL2, BTK, PI3K, and mTOR. These agents represent mechanistically distinct approaches being combined to improve outcomes in Richter transformation.
Ijms 26 08747 g002
Table 1. Incidence of Richter Transformation in Different Studies.
Table 1. Incidence of Richter Transformation in Different Studies.
Study (Year)SettingRT Incidence
MDACC
(1975 to 2005) [27]
CIT era, N = 3986
Retrospective
5.1%
GCLLSG
(1999 to 2016) [28]
CIT era, N = 2975
Pooled analysis of trials
2–10%
Nationwide Danish
(2008 to 2016) [19]
Retrospective
N = 3772
2.6%
DLBCL-RT
SEER registry [29]Retrospective
N = 74,166
0.7%
RESONATE [30]
(2012 to 2018 follow-up)
Phase 3, RCT, r/r CLL
Ibrutinib, n = 195
5.1%
RESONATE-2 [31]
(2013 to 2018 follow-up)
Phase 3, RCT,
Ibrutinib, n = 136
1.5%
MURANO [32]
(2014 to 2015)
Phase 3, RCT, r/r CLL
Venetoclax + Rituximab, n = 194
Bendamustine + Rituximab, n = 195
V + R, 3.1%
B + R, 2.6%
Abbreviations: CIT, chemoimmunotherapy; R/R, relapsed/refractory; RT, Richter transformation.
Table 2. Comparison of Therapeutic Strategies in RT.
Table 2. Comparison of Therapeutic Strategies in RT.
Therapy ProsConsComments
Traditional standard therapies
ChemoimmunotherapyBroad clinical accessibility
Induces rapid cytoreduction
Established use in standard practice
Low CR rate (~20–30%)
Median OS of 6–12 months
High frequency of relapse
Historically used as front-line therapy
Suboptimal for durable disease control
Often serves as induction prior to other approaches
Allo-HSCTOnly potentially curative approach
Enables long-term remissions (>3 years)
Leverages graft-versus-lymphoma effect
High TRM (up to 25%)
Restricted to transplant-eligible patients
Requires disease CR prior to allo-HSCT
Optimal for younger, fit patients in remission
Early referrals
Reduced-intensity conditioning expand eligibility
Novel approaches
BTK InhibitorsOral administration
Active in a subset of RT cases
Favorable tolerability
Modest ORR (~38–58%)
Short PFS (3–4 months)
Common resistance and early relapses
Frequently used as salvage or bridging therapy
Combination strategies may enhance efficacy
Limited clinical benefit as monotherapy
BCL2 InhibitorsEncouraging in combination regimens
ORR up to 90%
Chemotherapy-free options
Limited efficacy as monotherapy
High relapse rates with single-agent use
Potential risk of tumor lysis syndrome
Most effective in combination regimens
Commonly utilized as a bridge to allo-HSCT or CAR T-cell therapy
CAR T-cell TherapyHigh ORR (63–65%)
Achieve MRD-negative CR in some cases
Demonstrated efficacy in off-label use
Short DOR
Limited accessibility
T-cell dysfunction and exhaustion
Risk of immune-related toxicities
Promising option for chemo-refractory RT
Effective as a bridge to transplant in responders
Potential for enhanced efficacy in combination strategies
Bispecific AntibodiesOff-the-shelf availability
Repeat dosing for sustained control
ORR of 40–50% reported in RT
Manageable toxic profile
Long-term DOR remains uncertain
Limited data available
Optimal sequencing with CAR T-cell therapy or allo-HSCT is not yet defined
An emerging option, especially for post-CAR T relapses or patients ineligible for cellular therapy
Subcutaneous administration enhances accessibility and treatment adherence
Abbreviations: Allo-HSCT, allogeneic hematopoietic stem cell transplantation; BCL2, B-cell lymphoma 2; BTK, Bruton’s tyrosine kinase; CAR T, chimeric antigen receptor T cell; CR, complete response; MRD, minimal residual disease; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; RT, Richter transformation.
Table 3. Key Considerations and Clinical Challenges of CAR T-cell Therapy in Patients with RT.
Table 3. Key Considerations and Clinical Challenges of CAR T-cell Therapy in Patients with RT.
ConsiderationsComments
Aggressive
Disease
RT often harbors TP53 mutations, complex karyotype, and unmutated IGHV, all of which predict poor outcomes with conventional therapy.
Manufacturing BarriersRapidly progressing RT necessitates bridging therapy, which may impair CAR T-cell fitness.
Lymphodepleting regimens before infusion must be cautiously managed to avoid toxicity.
ToxicityCAR T-cell related adverse events like CRS and ICANS are prominent and more severe in RT patients due to high tumor burden.
Infection risk is elevated due to heavy pretreatment and aggressive disease.
On-Target,
Off-Tumor
Toxicity and
CD19 Escape
Loss of CD19 antigen expression can lead to CAR T-cell resistance.
RT cells may show heterogeneous CD19 expression, complicating targeting.
Limited Data RT patients are often excluded from most pivotal CAR T-cell trials.
Patient FactorsRT patients are often older and frailer, with prior toxicities from CLL therapies, limiting eligibility for CAR-T.
Functional status and disease kinetics must guide decision-making.
Abbreviations: CAR-T, chimeric antigen receptor T cell; CLL, chronic lymphocytic leukemia; CRS, cytokine release syndrome; ICANS, immune effector cell-associated neurotoxicity syndrome; IGHV, immunoglobulin heavy chain variable region; RT, Richter transformation; TP53, tumor protein p53.
Table 4. Reported Outcomes of Anti-CD19 CAR T-Cell Therapies in Richter Transformation.
Table 4. Reported Outcomes of Anti-CD19 CAR T-Cell Therapies in Richter Transformation.
ReferencePatient
Number
Prior
Treatment Lines
(Median)
ORR/CR (%)PFS/OS (mo)
Benjamini O et al. [81]30
(Axi-cel, n = 4; Tisa-cel, n = 7; Liso-cel, n = 3, POC anti-CD19 CAR T, n = 16)
2 (0–7)57/404.3/9.9
Kittai AS et al. [82]69
(Axi-cel, n = 44; Tisa-cel, n = 17, Liso-cel, n = 7; Brexu-cel, n = 1)
2 (0–7)63/464.7/8.5
Beyar-Katz, O. et al. [83]54
(Axi-cel, n = 4; Tisa-cel, n = 20; Liso-cel, n = 1, POC anti-CD19 CAR T, n = 29)
2 (0–8)65/4612-m PFS:41%/14.4
Kittai AS et al. [84] Axi-cel, n = 84100/63NR/NR
Abramson, J.S. et al. [85,86]Liso-cel, n = 4NR50/25NR/NR
Axi-cel: Axicabtagene ciloleucel; Brexu-cel: Brexucabtagene autoleucel; CAR: Chimeric antigen receptor; CR: Complete response; Liso-cel: lisocabtagene maraleucel; mo: month; NR: Not reported; ORR: Overall response rate; OS: Overall survival; PFS: Progression-free survival; POC: Point-of-care; Tisa-cel: Tisagenlecleucel.
Table 5. Comparison of Bispecific Antibody Therapies in RT.
Table 5. Comparison of Bispecific Antibody Therapies in RT.
Bispecific AntibodyTrial PhasePatient NumbersORR/CR (%)PFS, Median (mo)
Epcoritamab [101]I/II3550/3512.8
Mosuntuzumab [102]I/II2040/20NR
Both studies do not report overall survival. Abbreviations: CR, complete response; mo, months; NR, not reported; ORR, overall response rate; PFS, progression-free survival.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lin, Z.-C.; Chan, M.-J.; Jaing, T.-H.; Lin, T.-L.; Hung, Y.-S.; Su, Y.-J. Richter Transformation in Chronic Lymphocytic Leukemia: Current Treatment Challenges and Evolving Therapies. Int. J. Mol. Sci. 2025, 26, 8747. https://doi.org/10.3390/ijms26178747

AMA Style

Lin Z-C, Chan M-J, Jaing T-H, Lin T-L, Hung Y-S, Su Y-J. Richter Transformation in Chronic Lymphocytic Leukemia: Current Treatment Challenges and Evolving Therapies. International Journal of Molecular Sciences. 2025; 26(17):8747. https://doi.org/10.3390/ijms26178747

Chicago/Turabian Style

Lin, Zi-Chi, Ming-Jen Chan, Tang-Her Jaing, Tung-Liang Lin, Yu-Shin Hung, and Yi-Jiun Su. 2025. "Richter Transformation in Chronic Lymphocytic Leukemia: Current Treatment Challenges and Evolving Therapies" International Journal of Molecular Sciences 26, no. 17: 8747. https://doi.org/10.3390/ijms26178747

APA Style

Lin, Z.-C., Chan, M.-J., Jaing, T.-H., Lin, T.-L., Hung, Y.-S., & Su, Y.-J. (2025). Richter Transformation in Chronic Lymphocytic Leukemia: Current Treatment Challenges and Evolving Therapies. International Journal of Molecular Sciences, 26(17), 8747. https://doi.org/10.3390/ijms26178747

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop