The Challenge of Indefinite Therapy in Chronic Lymphocytic Leukemia
The landscape of Chronic Lymphocytic Leukemia (CLL) treatment has been fundamentally altered by the advent of Bruton tyrosine kinase (BTK) inhibitors. Ibrutinib, the first-in-class BTK inhibitor, shifted the paradigm from intensive chemoimmunotherapy to targeted oral treatment. While ibrutinib significantly improves progression-free and overall survival, it rarely achieves complete response (CR) or undetectable measurable residual disease (uMRD). Consequently, patients typically remain on ibrutinib indefinitely, leading to concerns regarding long-term toxicity, financial burden, and the eventual development of resistance mutations in BTK or PLCG2. The clinical community has therefore sought ‘finite’ therapy options that can induce deep, durable remissions allowing for treatment discontinuation.
The Rationale for BAFF Receptor Targeting
One promising strategy involves targeting the B-cell-activating factor receptor (BAFF-R), which is essential for the survival and maturation of B cells. Ianalumab (VAY736) is a novel, humanized, Fc-engineered monoclonal antibody that targets BAFF-R. It exerts its antitumor effect through two primary mechanisms: direct blockade of BAFF-R signaling and enhanced antibody-dependent cellular cytotoxicity (ADCC) mediated by natural killer (NK) cells. Preclinical models suggested that the combination of ianalumab and ibrutinib could synergistically reduce tumor burden and overcome the limitations of BTK inhibition alone.
Study Design and Methodology
The Phase Ib trial (NCT03400176) was an open-label, dose-escalation and expansion study designed to evaluate the safety, tolerability, and preliminary activity of ianalumab in combination with ibrutinib. The study enrolled 39 patients with CLL who had been on ibrutinib for at least 12 months but had not achieved a CR, or those who had developed evidence of resistance.
Patient Population and Dosing
The cohort included 15 patients in the dose-escalation phase and 24 in the expansion phase. Ianalumab was administered intravenously once every two weeks for up to eight cycles of 28 days. In the escalation phase, doses ranged from 0.3 mg/kg to 9.0 mg/kg; the recommended expansion dose was determined to be 3.0 mg/kg. All patients continued their standard daily dose of 420 mg ibrutinib. The primary endpoints were safety and the determination of the recommended dose, with secondary endpoints focusing on antitumor activity and the feasibility of ibrutinib discontinuation.
Key Findings: Safety and Tolerability
The combination of ianalumab and ibrutinib was found to be generally well tolerated, with no dose-limiting toxicities observed during the escalation phase.
Adverse Event Profile
Grade 3 or higher adverse events (AEs) occurred in 16 patients (41.0%). However, only nine of these patients (23.1%) had AEs that were considered treatment-related. Common toxicities were consistent with the known profiles of both agents, including infusion-related reactions (mostly Grade 1-2) and cytopenias. Importantly, there were no on-treatment deaths. One death occurred during the post-treatment follow-up period due to COVID-19, highlighting the ongoing vulnerability of this patient population rather than a direct drug-related toxicity.
Efficacy: Deep Molecular Responses
The efficacy data from this study were particularly striking, especially regarding the depth of response. At the end of the treatment period (Cycle 9), 38.5% of the total cohort achieved a CR or CR with incomplete marrow recovery (CRi).
Achieving uMRD
Perhaps the most significant finding was the rate of undetectable measurable residual disease (uMRD). On day 1 of Cycle 9, 17 patients (43.6%) achieved uMRD in the peripheral blood or bone marrow. This is a substantial improvement over historical data for ibrutinib monotherapy, where uMRD rates are typically in the single digits even after years of continuous treatment. The achievement of uMRD is increasingly recognized as a surrogate marker for long-term survival and is a prerequisite for successful therapy discontinuation.
Breaking the Cycle: Successful Treatment Discontinuation
The most clinically impactful result of the study was the ability of patients to stop ibrutinib. Of the 39 patients treated, 17 (43.6%) discontinued ibrutinib at or after Cycle 9, Day 1.
Durable Off-Therapy Remissions
These patients remained off therapy for a median duration ranging from 12.1 to 24.5 months. This ‘treatment holiday’ or treatment-free remission (TFR) represents a major quality-of-life improvement for patients who would otherwise face lifelong medication. The ability to achieve finite therapy with a combination of a BTK inhibitor and a BAFF-R antibody could redefine the standard of care for patients who do not achieve optimal responses on monotherapy.
Mechanistic Insights: NK and T-Cell Activation
The study also provided valuable biomarker data to explain the clinical success. Preliminary RNA sequencing and flow cytometry data suggested that ianalumab does more than just deplete B cells.
Immunomodulatory Effects
The researchers observed evidence of both NK-cell and T-cell activation following ianalumab administration. The Fc-engineering of ianalumab likely enhances ADCC, while the reduction in tumor burden may relieve some of the T-cell exhaustion typically seen in CLL. This dual action—direct cytotoxicity and immune microenvironment modulation—appears to be key to eliminating the residual CLL clones that survive ibrutinib treatment alone.
Expert Commentary and Clinical Implications
The results of this Phase Ib study are highly encouraging. For years, the ‘ibrutinib wall’—the plateau where patients maintain stable disease but harbor persistent residual leukemia—has been a hurdle in CLL management. By adding ianalumab, clinicians may have found a way to bridge the gap between stable disease and true molecular remission.
Strengths and Limitations
The primary strength of this study is the high rate of uMRD and the successful discontinuation of ibrutinib in a significant subset of patients. However, as a Phase Ib study, the sample size is relatively small. Long-term follow-up will be essential to determine the durability of these remissions and whether patients who eventually relapse remain sensitive to BTK inhibitors. The study also highlights the importance of patient selection, as those with specific resistance mutations may respond differently to the combination.
Conclusion
The addition of ianalumab to ibrutinib is a safe and highly effective strategy for deepening responses in CLL. With 43.6% of patients achieving uMRD and successfully entering a period of treatment-free remission, this combination therapy offers a potential path away from indefinite treatment. These findings strongly support the continued evaluation of ianalumab in larger, randomized clinical trials and suggest that targeting the BAFF receptor may be a vital component of future finite-duration treatment regimens in CLL.
Funding and Clinical Trial Information
This research was supported by Novartis Pharmaceuticals Corporation. The clinical trial registration number is NCT03400176.
References
1. Rogers KA, Yan P, Flinn IW, et al. Addition of Ianalumab (VAY736) to Ibrutinib in Patients with Chronic Lymphocytic Leukemia on Ibrutinib Therapy: Results from a Phase Ib Study. Clin Cancer Res. 2025;31(24):5145-5158. doi:10.1158/1078-0432.CCR-25-0210.
2. Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N Engl J Med. 2015;373(25):2425-2437.
3. Touzeau C, Le Gouill S. How I treat CLL with BTK inhibitors. Blood. 2021;137(13):1710-1717.
4. Woyach JA, Ruppert AS, Heerema NA, et al. Ibrutinib Regimens versus Chemoimmunotherapy in Older Patients with Untreated CLL. N Engl J Med. 2018;379(26):2517-2528.

