1,112
Views
0
CrossRef citations to date
0
Altmetric
Review Article

Bruton’s tyrosine kinase (BTK) inhibitors for the treatment of primary central nervous system lymphoma (PCNSL): current progress and latest advances

, &
Received 10 Dec 2023, Accepted 18 Mar 2024, Published online: 10 Apr 2024

Abstract

The incidence of primary central nervous system lymphoma (PCNSL) has steadily increased, particularly in elderly patients. Although highly responsive to first-line chemotherapy and radiotherapy, approximately 50% of patients relapse or become refractory within 1 year. Prognosis following relapse is dismal and no standard salvage therapy exists. Bruton’s tyrosine kinase (BTK), a key regulator of the B-cell receptor (BCR) pathway, has emerged as a promising therapeutic target. The first BTK inhibitor ibrutinib has been evaluated in the relapsed/refractory PCNSL setting, with overall response rates of 51.9%–89.0% and median progression-free survival of 4.6–4.8 months. However, ibrutinib inhibits several kinases in addition to BTK, leading to off-target effects. Second-generation BTK inhibitors have since been developed, which afford greater selectivity for BTK and fewer off-target effects. We review current practices in the diagnosis and evaluation of PCNSL, as well as clinical trials of BTK inhibitors in PCNSL and future developments in PCNSL treatment.

Introduction

Primary central nervous system lymphoma (PCNSL) is a highly aggressive non-Hodgkin lymphoma (NHL) restricted to the brain, meninges, spinal cord, and eyes and is histopathologically predominantly classified as diffuse large B-cell lymphoma (DLBCL) (90%). In immunocompetent patients, advancing age is a primary risk factor for PCNSL, with a median age at diagnosis of 65 years, and approximately one-third of patients aged ≥70 years [Citation1]. PCNSL is considered a relatively rare tumor, accounting for 4%–6% of all extranodal lymphomas in the USA [Citation2,Citation3]. However, epidemiological data shows that the incidence of PCNSL has been steadily rising in recent decades in parallel with the burgeoning aging population. In an analysis of data from the Central Brain Tumor Registry of the USA and the Surveillance, Epidemiology, and End Results registry, the incidence of PCNSL rose from 0.1 patients per 100,000/years in the 1970s to 0.4 patients per 100,000/years in 2013 [Citation4]. Of these, elderly patients were disproportionately affected, with an incidence rate of 4.32 per 100,000/years in patients aged 70–79 years in the USA [Citation4].

High-dose methotrexate (HD-MTX)-based regimens are considered the standard of care for newly diagnosed PCNSL [Citation5]. Although PCNSL is highly responsive to chemotherapy and radiation therapy in the first-line setting [Citation5], median time to relapse is 10–18 months and most relapses occur within 2 years [Citation3]. The prognosis for patients with relapsed/refractory (r/r) PCNSL is dismal, with a median overall survival (OS) of 8.4 months despite salvage therapy [Citation6]. Furthermore, many elderly patients are not able to tolerate intensive treatments due to poor performance status, impaired renal function (CCr < 50 ml/min), and other comorbidities, and are particularly susceptible to treatment-associated toxicity [Citation7]. Whole-brain radiotherapy (WBRT) has traditionally been used as consolidation following HD-MTX-based chemotherapy but is associated with a significant risk of permanent and irreversible neurotoxicity, particularly in patients aged ≥60 years [Citation8].

Bruton’s tyrosine kinase (BTK) is a key regulator of the B-cell receptor (BCR) pathway and a promising target in the treatment of lymphomas with constitutive activation of the nuclear factor kappa β (NF-κB) pathway. Due to the central role of the BCR signaling pathway in B-cell survival and proliferation, BTK represents an attractive molecular target for the treatment of B-cell NHL, and PCNSL in particular. Characterized by DLBCL, PCNSL exhibits diverse molecular features, including alterations in key genes associated with B-cell lymphomas. MYD88 (primarily L265P) and CD79B (primarily Y196) are the most frequently mutated genes in DLBCL [Citation9], which lead to constitutive activation of the BCR pathway, including NF-κβ and downstream signals responsible for the activation, proliferation, and survival of B-cell lymphomas () [Citation9,Citation10]. Over 75% of patients with PCNSL possess mutations in one or both of these genes [Citation11]. In recent genomic analyses, PCNSL also frequently exhibits 9p24.1/PD-L1 (programmed cell death-ligand 1)/PD-L2 (programmed cell death-ligand 2) copy number alterations and translocations, which are likely genetic bases of immune evasion. In a small case series of 21 patients with PCNSL, 67% had 9p24.1/PD-L1/PD-L2 copy gain and copy number-associated increased expression of the 2 ligands [Citation12]. Another study reported that only 4% of PCNSL tissue specimens had PD-L1-positive tumor cells; however, 52% of the samples had PD-L1-positive cells in the tumor microenvironment [Citation13]. Mutations in TP53, PIM1, and IRF4 are also frequently reported in PCNSL and may play a role in disease development and progression [Citation14]. However, it should be noted that no known molecular signature to predict therapy failure/resistance.

Figure 1. Overview of the BTK signaling pathway.

BTK: Bruton’s tyrosine kinase.

Figure 1. Overview of the BTK signaling pathway.BTK: Bruton’s tyrosine kinase.

In this review, we summarize recent progress and the latest advances in the development of BTK inhibitors for the treatment of PCNSL.

BTK inhibitors for the treatment of PCNSL

To date, five BTK inhibitors – ibrutinib, acalabrutinib, zanubrutinib, tirabrutinib, and orelabrutinib – have been approved for the treatment of hematological malignancies, all of which are covalent and irreversible inhibitors of BTK activity (). BTK inhibitors act by binding to the active site of BTK, cysteine-481 (Cys-481) to inhibit the overexpression and phosphorylation of tyrosine-223 and aberrant activation of the NF-κβ pathway.

Table 1. Summary of approved BTK inhibitors.

BTK kinase selectivity and safety profile of existing BTK inhibitors

Each BTK inhibitor possesses its own unique safety profile, with differences in kinase selectivity believed to be largely responsible for the variations observed in adverse event (AE) profiles [Citation15]. In addition to BTK, ibrutinib has been shown to irreversibly inhibit the activity of several important kinases, including epidermal growth factor receptor (EGFR), which is associated with severe skin toxicities [Citation16], IL-2-inducible T-cell kinase (ITK), an important regulator of natural killer cell-mediated cytotoxicity [Citation17], and other members in the tyrosine kinase expressed in hepatocellular carcinoma (TEC) family of kinases (such as bone marrow tyrosine kinase gene on chromosome X [BMX]), which are involved in diverse biological processes related to cell survival and differentiation [Citation18]. In contrast, in vitro kinase assays demonstrate that the second-generation BTK inhibitors, tirabrutinib, acalabrutinib, and zanubrutinib, possess more selective kinase binding compared with ibrutinib and thus fewer off-target effects () [Citation19]. In a study evaluating half-maximal inhibitory concentrations (IC50) of BTK inhibitors against BTK and 13 kinases, the IC50 of the second-generation inhibitors was considerably higher than ibrutinib against all kinases tested, and there were 5 kinases that ibrutinib inhibited to a similar degree as BTK (IC50 within 10-fold of BTK) compared with only 2–3 kinases with the second-generation BTK inhibitors () [Citation19].

Figure 2. Kinome profiling using the KINOMEscan assay for the first- and second-generation BTK inhibitorsa.

BTK: Bruton’s tyrosine kinase.

aReproduced from Kozaki et al. [Citation19].

Figure 2. Kinome profiling using the KINOMEscan assay for the first- and second-generation BTK inhibitorsa.BTK: Bruton’s tyrosine kinase.aReproduced from Kozaki et al. [Citation19].

Figure 3. MRI image of patient who responded to BTK inhibitors (tirabrutinib, ONO-4059-02 trial) in PCNSL.

MRI: magnetic resonance imaging; PCNSL: primary central nervous system lymphoma.

Figure 3. MRI image of patient who responded to BTK inhibitors (tirabrutinib, ONO-4059-02 trial) in PCNSL.MRI: magnetic resonance imaging; PCNSL: primary central nervous system lymphoma.

Table 2. Selectivity of first- and second-generation BTK inhibitors targeting 14 kinases.

BTK inhibitor common AEs

Ibrutinib is generally well tolerated but carries a clinically significant risk of cardiovascular events, bleeding, infection, diarrhea, and dermatological AEs, which often lead to dose reductions and treatment discontinuation [Citation20]. Cardiovascular AEs are perhaps the most notable off-target effect associated with ibrutinib treatment, with an increased risk of new or worsening hypertension (78.3%), atrial fibrillation (AF) (4%–13%), and minor (16%–55%) and major (2%–6%) bleeding reported across clinical studies [Citation20–22]. In contrast, fewer cardiovascular events are reported with the second-generation BTK inhibitors, acalabrutinib (hypertension: 9%; AF: 3%–7%; major bleeding: 6%) [Citation23,Citation24], zanubrutinib (hypertension: 12%; AF: 3%; major bleeding: 4%) [Citation25], and tirabrutinib (hypertension: 0%–3.7%; AF: 0%–7%; major bleeding: 0%) [Citation15,Citation26].

Compared with second-generation BTK inhibitors, ibrutinib is a more potent inhibitor of human epidermal growth factor 2 (HER2) and 4 (HER4) [Citation15], and inhibition of HER2/HER4 expression in cardiomyocytes by ibrutinib is widely considered to be the mechanism responsible for the increased incidence of cardiovascular events observed [Citation15]. In a review of studies evaluating kinase activity of BTK inhibitors, ibrutinib was found to inhibit all kinases implicated in causing cardiovascular AEs, namely HER2, HER4, and TEC [Citation15]. Conversely, acalabrutinib was found to inhibit HER4, and only slightly TEC, but not HER2, zanubrutinib inhibited TEC and HER4, but not HER2, and tirabrutinib inhibited TEC but not HER2 or HER4. More recently, C-terminal Src kinase (CSK) has emerged as another potential molecular target underlying AF after it was identified on a short list of candidate kinases, and cardiac symptoms were reproduced in cardiac-specific CSK knockout mice [Citation27]. Consistent with the low incidence of cardiovascular events, zanubrutinib demonstrates less inhibition of CSK compared with ibrutinib and acalabrutinib does not inhibit Src family kinases [Citation28,Citation29]. The cumulative incidence of new or worsened hypertension in patients treated with ibrutinib at 1.8 months was also reported to be 50%, and the cardiovascular effects of long-term use should be considered [Citation22].

Dermatological reactions and diarrhea are among the most commonly experienced AEs with ibrutinib, most of which are grade 1–2 in severity and occur within the first 6 months to 1 year of treatment [Citation30,Citation31]. In a recent review evaluating the relative incidence of dermatological AEs in phase 2/3 studies with BTK inhibitors, rash occurred at a similar rate between patients treated with ibrutinib (13%–27%; grade ≥3: 0%–3%), acalabrutinib (15%–18%), and zanubrutinib (13%–18%) [Citation31]. However, in the Phase 1/2 trial of tirabrutinib in Japanese patients with r/r PCNSL (ONO-4059-02), a relatively high incidence of skin disorders was reported, including rash (31.8%; grade ≥3: 2.3%), erythema multiforme (11.4%; grade ≥3: 6.8%), drug eruption (9.1%; grade ≥3: 4.5%), and maculo-papular rash (6.8%; grade ≥3: 2.3%) [Citation32]. Diarrhea is reported in approximately 50% of patients during ibrutinib treatment (grade ≥3: 5%–7%) [Citation33,Citation34], which is a similar frequency to that reported during acalabrutinib (35%–52%), and tirabrutinib (25%–44%) treatment, although severity is lower with the newer agents (grade ≥3 events: 0%–5% vs. 0%–7%, respectively) [Citation35–38].

Dermatological reactions and diarrhea are generally thought to occur with BTK inhibitors as a result of off-target inhibition of EGFR. Gastrointestinal and dermatological AEs are well-recognized class effects with EGFR inhibitors [Citation39], and cutaneous manifestations with ibrutinib closely resemble those experienced with EGFR inhibitors [Citation40]. The incidence of dermatological reactions and diarrhea reported with second-generation BTK inhibitors, which have a relatively low potency for EGFR inhibition, suggest that other mechanisms distinct from EGFR may be at play and warrant further investigation.

Although considered to be less immunosuppressive than conventional immunochemotherapy, several cases of invasive fungal infections have been reported in clinical trials of BTK inhibitors in PCNSL [Citation32,Citation41]. Ghez et al. reported on 33 patients who developed invasive fungal infections during ibrutinib treatment, the majority of which were invasive aspergillosis [Citation42]. However, the majority of patients also had at least one additional factor that increased their risk for fungal infections, including chemotherapy within the last 6 months, neutropenia, or corticosteroid use. Second-generation BTK inhibitors such as acalabrutinib and zanubrutinib possess greater binding selectivity and fewer off-target effects compared to ibrutinib but disseminated fungal infections have still been reported, albeit less frequently [Citation32,Citation41]. In a Phase 1/2 study of tirabrutinib in R/R PCNSL, grade ≥3 bronchopulmonary aspergillosis, pneumonia, and Pneumocystis jirovecii pneumonia occurred in 1 of 44 patients (2.3%) each [Citation32]. In a phase 2 study of orelabrutinib in combination with an anti-PD-1 monoclonal antibody in R/R PCNSL (n = 13), 1 patient experienced a grade 3 AE of interstitial pneumonitis-related Pneumocystis jirovecii infection [Citation41]. These findings indicate that susceptibility to invasive fungal infection and pneumocystis pneumonia is not merely a consequence of off-target effects on T cell response and suggests a greater role of B-cell pathways in antifungal immunity, although the exact mechanism/s by which BTK inhibitors raise the risk of these events are still under investigation. Given that corticosteroid use is common in the treatment of PCNSL and the risk of invasive fungal infections and pneumocystis pneumonia appears higher in this cohort compared with other non-central nervous system (CNS) lymphomas and chronic lymphocytic leukemia (CLL), caution is warranted in those patients.

Penetration of BTK inhibitors across the blood–brain barrier

Given that PCNSL is restricted to the brain, eyes, meninges, and other structures of the CNS, drugs that are capable of crossing the blood–brain barrier (BBB) to deliver sufficient cytotoxic doses to malignant cells and tissues in the CNS are necessary to achieve durable responses. However, the vast majority of chemotherapeutic agents that are effective in other systemic lymphomas have limited penetration of the BBB. For example, standard doses of MTX do not cross the BBB, necessitating administration of high doses and/or rapid infusion, which increase the risk of systemic and neurological toxicity.

BTK inhibitors are small molecules that have been shown to readily cross the BBB, with a cerebrospinal fluid (CSF) to total plasma concentration ratio of 1%–7% for ibrutinib [Citation43], 0.61%–5.83% for zanubrutinib [Citation44], and 13%–18% for tirabrutinib [Citation32]. The timing of pharmacokinetics (PK) evaluations differed (2–4 h ibrutinib/zanubrtinitb, 24 h tirabrutinib), limiting direct comparison.

Drug–drug interaction with methotrexate

A lack of high-quality data exists regarding a potential interaction between MTX and BTK inhibitors in the literature. However, when BTK inhibitors are used in combination with high-dose MTX-based therapy, pharmacokinetic interactions via CYP3A4 are a concern and should be considered with caution [Citation45,Citation46]. In a phase 1b study evaluating ibrutinib in combination with HD-MTX and rituximab (RTX) in patients with r/r CNS lymphoma, the ibrutinib dose was reduced due to a drug interaction with the CYP3A inhibitor, amlodipine, which was initiated to control AF [Citation47]. However, no dose-limiting toxicities (DLTs), treatment-related deaths, or cases of aspergillosis were observed and all grade 4 AEs were seen in patients treated with RTX, HD-MTX, and ibrutinib (840 mg, n = 2; 560 mg, n = 2). Similarly, most (62%) grade 3 AEs were seen in patients treated with this combination. Using a rat model of collagen-induced arthritis, potential drug–drug interactions between HM71224, a BTK inhibitor, and MTX were explored. HM71224 in combination with MTX was found to decrease the arthritis score, bone erosion, synovitis, and cartilage degradation without apparent interaction [Citation48].

Efficacy and safety of BTK inhibitors for PCNSL in clinical trials

Ibrutinib

To date, two prospective clinical studies have been conducted evaluating the efficacy and safety of ibrutinib monotherapy for the treatment of r/r PCNSL (). In an open-label, Phase 1 dose escalation study evaluating 560 mg and 840 mg of ibrutinib, 77% (10/13) achieved a clinical response (5 each with a complete response [CR] and partial response [PR]), and the median progression-free survival (PFS) was 4.6 months [Citation9]. In a phase 2 study of 52 patients receiving ibrutinib 560 mg once daily for r/r PCNSL and primary vitreoretinal lymphoma, a CR rate of 19% (10/52 patients) and a PR rate of 33% (17/52 patients) was achieved, and the median PFS was 4.8 months [Citation49]. Grommes et al. reported that while PCNSL patients with MYD88/CD79B co-mutations that activate BCR signaling were expected to respond better to ibrutinib than wild-type patients, there was no apparent response differentiation between patients with or without the co-mutations [Citation9]. In the phase 1 study, hyperglycemia (95%), thrombocytopenia (70%), anemia (70%), and hypertriglyceridemia (70%) were the most common AEs, however, in the above two studies three patients experienced pulmonary aspergillosis, with a favorable (n = 1) and fatal (n = 2) outcome [Citation49]. Based on the results of the aforementioned studies, ibrutinib monotherapy is listed in the NCCN guidelines as a treatment option for r/r PCNSL [Citation50].

Table 3. Clinical trials of BTK inhibitors for r/r PCNSL.

More recently, ibrutinib has been investigated in combination with other therapies for the treatment of r/r PCNSL (). In a phase 1 study of ibrutinib in combination with HD-MTX and RTX in patients with r/r CNS lymphoma, an overall response rate (ORR) of 89% (8/9 patients) was achieved and the median PFS was not reached in the PCNSL cohort [Citation47]. The most common AEs were anemia, thrombocytopenia, alanine aminotransferase/aspartate aminotransferase elevation, and lymphopenia. No DLTs, treatment-related death, or cases of aspergillosis were observed. A phase 1b study has also been conducted of ibrutinib monotherapy followed by ibrutinib plus chemotherapy (DA-TEDDi-R) in patients with PCNSL and CD79B and/or MYD88 mutations, in which 18 patients were treated with ibrutinib monotherapy, 16 patients started TEDDi-R treatment and 14 were evaluated [Citation11]. A total of 94% of patients showed tumor reductions with ibrutinib alone, and 12 (86%) achieved a complete remission with DA-TEDDi-R. However, the combination of ibrutinib with chemotherapeutic agents was associated with invasive aspergillosis in 7/18 patients (39%), three of which were related, one was probably related, and three were possibly related.

Tirabrutinib

A phase 1/2 study (ONO-4059-02) has been conducted of tirabrutinib in 44 Japanese patients with r/r PCNSL using three dosing regimens (320 mg, 480 mg, and 480 mg in the fasting state) () [Citation32]. During the 9-month follow-up period, the ORR was 63.6% (28/44 patients) in the overall population, and 60.0% (12/20 patients) in the 320 mg group, 100% (7/7 patients) in the 480 mg group, and 52.9% (9/17 patients) in the 480 mg fasted group. CR was achieved in 9.1% (4/44 patients), unconfirmed complete response (CRu) in 25% (11/44 patients), and PR in 29.5% (13/44 patients) of patients. shows the MRI image of a patient with PCNSL who responded to tirabrutinib in the ONO-4059-02 trial. No notable differences in ORR were observed between patients receiving tirabrutinib with gene mutations versus those without gene mutations in the phase 1/2 study (ONO-4059-02), including CARD11 (58.8% [10/17] vs. 66.7% [18/27]), MYD88 (59.4% [19/32] vs. 75.0% [9/12]), and CD79B (50.0% [9/18] vs. 73.1% [19/26]) mutations [Citation32]. The median PFS was 2.9 months (95% confidence interval [CI]: 1.8–11.1) in the overall population, 2.1 months (95% CI: 1.8–not evaluable [NE]) in the 320 mg group, 11.1 months (95% CI: 1.4–NE) in the 480 mg group, and 5.8 months (95% CI: 1.0–5.8) in the 480 mg fasted group. No DLTs were observed in this study. AEs occurred in 38 of the 44 patients (all grades), and grade ≥3 AEs occurred in approximately 50% of patients. The most common AEs were skin disorders (31.8%), leukopenia (18.2%), and lymphopenia (15.9%), and the most common grade ≥3 AEs were neutropenia (9.1%), lymphopenia, leukopenia, and erythema multiforme (6.8% each). If administration of tirabrutinib was interrupted in response to a skin disorder, prophylactic use of antiallergic drugs or corticosteroids at the resumption of administration was considered [Citation32]. Skin disorders resolved after treatment with an antihistamine or corticosteroid, which facilitated continued tirabrutinib administration. There were no cardiovascular-related AEs of any grade, including AF or hypotension. The final 3-year follow-up data have now been published for the entire 44 patients [Citation52]. The ORR was 63.6%, including 36.4% of patients who achieved a CR, the median duration response was 9.2 months, and the median PFS was 2.9 months. No new grade 5 AEs nor new safety profiles were observed since the last data cutoff.

A prospective, noninterventional, multicenter, observational post-marketing all-case surveillance study (JRCT ID: jRCT2011210002) has since been conducted to evaluate adverse drug reactions (ADRs) and the ORR of tirabrutinib in r/r PCNSL in Japan [Citation53]. During an interim analysis (data cutoff date: 31 October 2020), 140 patients who received tirabrutinib treatment were analyzed for safety, and data from 127 of those patients were analyzed for effectiveness. The most common ADRs were skin and subcutaneous tissue disorders (30.7%), and no new safety concerns were identified compared with those reported in the phase 1/2 study. No instances of cardiovascular ADRs or grade 5 ADRs were observed. Furthermore, the ORR was 63.0% and the rate of CR or CRu response (37% each) was similar to that achieved in the 480 mg fasted group in the phase 1/2 ONO-4059-02 study [Citation32]. By the end of the 52 weeks, 73 patients (52.1%) were still alive, 46 (32.9%) had died, and the status of 21 (15.0%) was unknown.

Zanubrutinib

Zanubrutinib in combination with lenalidomide, temozolomide (TMZ), and RTX, with or without methotrexate (RLZT ± MTX) has been shown to be an effective and safe treatment option in the first-line setting for patients with PCNSL (). In a prospective, open-label study, the efficacy and safety of RLZT + MTX were compared against RLZT alone in newly diagnosed patients with PCNSL [Citation51]. At data cutoff (30 June 2022), 24 patients were enrolled in the study, including 12 patients each in the RLZT + MTX and RLZT groups, respectively. The ORR in the overall population was 79.2%, including 9 (37.5%) patients with CR and 10 (41.7%) patients with PR; 2 (8.3%) patients had SD, and 3 (12.5%) patients had progressive disease (PD). The ORR was 66.7% (CR: 16.7%; PR: 50%) in the RLZT + MTX group and 91.7% (CR: 58.3%; PR: 33.3%) in the RLZT group. Median PFS and OS were not achieved in the overall population, and the estimated 18-month OS and PFS were 95.8% and 78.2%, respectively. Overall, 50% of patients experienced hematologic toxicity. The incidence of non-hematologic toxicity was 50% and included gastrointestinal reactions, rash, and liver and kidney damage. Only one patient experienced grade ≥3 rash. However, pneumonia was reported in 16.7% (4/24) of patients.

Orelabrutinib

Preliminary results have recently been published from an ongoing prospective, multicenter, single-arm phase 2 study evaluating orelabrutinib with the programmed cell death protein 1 (PD-1) inhibitor, sintilimab, for r/r PCNSL [Citation41]. Thirteen patients were enrolled from March 2021 to January 2022, with a median follow-up of 7.0 (1.5–10.5) months. At data cutoff, the ORR was 61.5%; 4 (30.7%) achieved CR, 1 (7.7%) achieved CRu, and 3 (23.1%) achieved PR, and the estimated 1-year PFS rate was 67.7%. Ten patients completed 4 cycles of the combination regimen, while 3 patients discontinued treatment in the first 2 cycles due to PD. Aside from one grade 3 AE of interstitial pneumonitis-related Pneumocystis jirovecii infection, no other grade 3–4 hematological or non-hematological AEs were reported, and toxicities were mild.

Despite these promising results, it should be noted that cohorts of PCNSL patients are extremely challenging to collect given the relative rarity of the disease, and many of the clinical trials included ≤20 patients, thus making new interventions incredibly difficult to assess.

Future directions for BTK inhibitors

Future directions

Development status of BTK inhibitors for PCNSL

Currently, there are several BTK inhibitors in development for the treatment of PCNSL, and their use in combination with chemotherapy, molecular targeted drugs, and immune checkpoint inhibitors is also being explored (). Newer BTK inhibitors such as pirtobrutinib and vecabrutinib are also under investigation in hemato-oncology, with the possibility for expansion into PCNSL in the future ().

Table 4. Development status of BTK inhibitors for PCNSL.

Table 5. Next-generation BTK inhibitors.

Treatment of newly diagnosed treatment-naïve PCNSL

HD-MTX-based chemotherapy regimens are recommended as first-line treatment for newly diagnosed PCNSL. However, toxicity and the need for frequent hospitalizations are a concern. Initiation of BTK inhibitors prior to recurrence might further optimize patient outcomes (). It has been reported that synergistic effects can be expected when HD-MTX and BTK inhibitors are used in combination. Currently, ibrutinib is also being studied as an add-on chemotherapy regimen with RTX, MTX, procarbazine, and vincristine (R-MPV) to further improve first-line therapy for PCNSL (; NCT02315326). The results of the Phase 1b LOC-R01 study (NCT04446962) of ibrutinib added to R-MPV in France have been reported, with 3/11 evaluable patients achieving CR/CRu and 7/11 achieving PR in the ibrutinib group [Citation54]. A Phase 2 study of tirabrutinib in patients with r/r and untreated PCNSL is ongoing in the USA (PROSPECT study Part B). In patients with untreated PCNSL, the safety and efficacy of tirabrutinib added to either MTX, TMZ, and RTX or R-MPV is being evaluated (; NCT04947319). The efficacy of orelabrutinib, which is under development in China, is also being investigated in a Phase 2 study in combination with RTX and MTX for the treatment of newly diagnosed PCNCL (; NCT05549284). These results suggest that BTK inhibitors are an attractive add-on to existing chemotherapy regimens.

Secondary CNS lymphoma

Secondary CNS lymphoma (SCNSL) refers to lymphoma that has metastasized to the CNS during or after systemic therapy. Relapse outside of the CNS, including at extranodal sites, is observed in approximately 16% of patients [Citation55]. SCNSL, which is classified into DLBCL, is a highly malignant tumor like PCNSL and early diagnosis is therefore necessary to achieve optimal therapeutic effects and improved prognosis. There is no standard treatment for SCNSL and prognosis is extremely dismal (median OS of 1.5 months) [Citation56]. Conventional PCNSL treatment with MTX or cytarabine chemotherapy is the treatment strategy of choice for SCNSL but is often inactive against primary chemorefractory disease [Citation57]. As is the case with PCNSL, BTK inhibitors are also expected to demonstrate a therapeutic effect in SCNSL. In a phase 2 study in 44 patients with r/r CNS lymphoma (PCNSL; n = 29, SCNSL; n = 15), treatment with ibrutinib was associated with an ORR of 69% in patients with SCNSL [Citation58] which is in contrast to the less impressive efficacy in non-CNS lymphomas, where only 37% responded [Citation59]. That being said, the proportion of MYD88 mutations is lower in SCNSL than in PCNSL [Citation60], and further investigation is therefore required to evaluate whether a similar level of efficacy observed with PCNSL may also be achieved with SCNSL. Combination regimens utilizing BTK inhibitors and other novel agents currently being investigated for SCNSL are presented in .

Next-generation BTK inhibitors

First- and second-generation BTK-targeting agents have shown impressive clinical responses in PCNSL; however, reports detailing the development of acquired resistance to ibrutinib have been published and are of concern [Citation61]. Mutations in the active site of BTK have been identified as the main mechanism of acquired resistance to ibrutinib in CLL and mantle cell lymphoma (MCL), with the most common mutation sites being C481S/F/T/R, as well as less frequent variants at T474I/S/M, R490H, Q516Km, L528W, and V537I [Citation62]. However, the mechanism of resistance in PCNSL is not currently clear and remains speculative.

The rise of mutations that are refractory to current therapies has presented a need for new therapeutic approaches in PCNSL. To address the problem of acquired resistance, next-generation BTK inhibitors, which are potent inhibitors of BTK as well as different variants of the BTK C481 mutation, are being developed for use in patients with CLL and MCL refractory to first- or second-generation BTK inhibitors () [Citation63]. Pirtobrutinib, a highly selective reversible inhibitor that acts on both wild-type and C481 mutant BTK [Citation64], is currently being developed, as are dual BTK-based inhibitors designed to overcome acquired resistance. Development of a novel BTK inhibitor with efficacy in patients with PCNSL despite acquired resistance to first- and second-generation BTK inhibitors is eagerly anticipated [Citation65]. Recently, protein degraders have been developed that target and degrade the BTK protein for BTK inhibition, with several molecules in Phase 1 clinical trials for various B-cell malignancies (Beigene [NCT05006716]; Nurix [NCT04830137, NCT05131022]; Haisco [NCT04861779]) and promising preliminary efficacy observed.

Conclusion

Although PCNSL is highly responsive to chemotherapy and radiation therapy in the first-line setting, the recurrence rate remains unacceptably high and prognosis is extremely poor. New treatment strategies with novel mechanisms of action distinct from conventional therapies are therefore of interest. PCNSL is associated with constitutive activation of BCR signaling, and BTK plays an important role in oncogenic signal transduction downstream of BCR, making it an attractive molecular target. Following the efficacy of the first-generation BTK inhibitor ibrutinib for the treatment of PCNSL, second-generation BTK inhibitors tirabrutinib, zanubrutinib, and orelabrutinib, have subsequently been developed, which possess greater selectivity for BTK and thus fewer off-target effects. These agents have delivered impressive treatment responses in the relapsed/refractory setting with acceptable safety. The expansion of these BTK inhibitors to newly diagnosed PCNSL, and potentially SCNSL, and the development of next-generation BTK inhibitors are expected to revolutionize the treatment landscape for PCNSL.

Author contributions

CG developed the concept for the review and assisted in performing the literature search. All authors critically reviewed, edited, and revised the manuscript, and approved the final draft for submission.

Acknowledgments

All authors participated in the writing, editing, and critical revision for intellectual content, and approval of the final version of this manuscript. All authors met ICMJE authorship criteria and agreed to be accountable for all aspects of the work.

Disclosure statement

LRS: grant support from the National Institutes of Health (P30-CA008748). Support for manuscript preparation received from ONO. Advisory Board fees from BTG Speciality Pharmaceuticals. Consultation fees from BTG Speciality Pharmaceuticals and ONO. Research support from BTG. Patent pending for low-dose glucarpidase. LN: support for manuscript preparation received from ONO. Grant support from the Leukemia and Lymphoma Society and the National Institutes of Health. Royalties or license fees received from Wolters Kluwer (UpToDate Inc). Consultation fees from ONO, BraveBio, and Genmab. Payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events received from ONO. Support for attending meetings and/or travel received from ONO. Participation on a Data Safety Monitoring Board or Advisory Board from ONO and Kite/Gilead. CG: grant support from the National Institutes of Health (P30-CA008748). Consultation fees from BTG Speciality Pharmaceuticals, Roche, and ONO. Support for manuscript preparation received from ONO. Payment or honoraria for lectures, presentations, and speakers’ bureaus received from ONO. Support for attending meetings and/or travel received from ONO. Participation on an Advisory Board from ONO.

Additional information

Funding

Medical writing support was provided by Jordana Campbell, BSc, CMPP of inScience Communications, Springer Healthcare. This medical writing assistance was funded by ONO Pharmaceutical Co., Ltd.

References

  • van der Meulen M, Dinmohamed AG, Visser O, et al. Improved survival in primary central nervous system lymphoma up to age 70 only: a population-based study on incidence, primary treatment and survival in The Netherlands, 1989-2015. Leukemia. 2017;31(8):1822–1825. doi:10.1038/leu.2017.128
  • Villano JL, Koshy M, Shaikh H, et al. Age, gender, and racial differences in incidence and survival in primary CNS lymphoma. Br J Cancer. 2011;105(9):1414–1418. doi:10.1038/bjc.2011.357
  • Grommes C, DeAngelis LM. Primary CNS lymphoma. J Clin Oncol. 2017;35(21):2410–2418. doi:10.1200/JCO.2017.72.7602
  • Mendez JS, Ostrom QT, Gittleman H, et al. The elderly left behind-changes in survival trends of primary central nervous system lymphoma over the past 4 decades. Neuro Oncol. 2018;20(5):687–694. doi:10.1093/neuonc/nox187
  • Grommes C, Rubenstein JL, DeAngelis LM, et al. Comprehensive approach to diagnosis and treatment of newly diagnosed primary CNS lymphoma. Neuro Oncol. 2019;21(3):296–305. doi:10.1093/neuonc/noy192
  • Langner-Lemercier S, Houillier C, Soussain C, et al. Primary CNS lymphoma at first relapse/progression: characteristics, management, and outcome of 256 patients from the French LOC network. Neuro Oncol. 2016;18(9):1297–1303. doi:10.1093/neuonc/now033
  • Morales-Martinez A, Lozano-Sanchez F, Duran-Peña A, et al. Primary central nervous system lymphoma in elderly patients: management and perspectives. Cancers (Basel). 2021;13(14):3479. doi:10.3390/cancers13143479
  • Bataille B, Delwail V, Menet E, et al. Primary intracerebral malignant lymphoma: report of 248 cases. J Neurosurg. 2000;92(2):261–266. doi:10.3171/jns.2000.92.2.0261
  • Grommes C, Pastore A, Palaskas N, et al. Ibrutinib unmasks critical role of Bruton tyrosine kinase in primary CNS lymphoma. Cancer Discov. 2017;7(9):1018–1029. doi:10.1158/2159-8290.CD-17-0613
  • Tao K, Wang X, Tian X. Relapsed primary central nervous system lymphoma: current advances. Front Oncol. 2021;11:649789. doi:10.3389/fonc.2021.649789
  • Lionakis MS, Dunleavy K, Roschewski M, et al. Inhibition of B cell receptor signaling by ibrutinib in primary CNS lymphoma. Cancer Cell. 2017;31(6):833–843.e5. doi:10.1016/j.ccell.2017.04.012
  • Chapuy B, Roemer MGM, Stewart C, et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood. 2016;127(7):869–881. doi:10.1182/blood-2015-10-673236
  • Hayano A, Komohara Y, Takashima Y, et al. Programmed cell death ligand 1 expression in primary central nervous system lymphomas: a clinicopathological study. Anticancer Res. 2017;37:5655–5666.
  • Bruno A, Boisselier B, Labreche K, et al. Mutational analysis of primary central nervous system lymphoma. Oncotarget. 2014;5(13):5065–5075. doi:10.18632/oncotarget.2080
  • Estupiñán HY, Berglöf A, Zain R, et al. Comparative analysis of BTK inhibitors and mechanisms underlying adverse effects. Front Cell Dev Biol. 2021;9:630942. doi:10.3389/fcell.2021.630942
  • Holcmann M, Sibilia M. Mechanisms underlying skin disorders induced by EGFR inhibitors. Mol Cell Oncol. 2015;2(4):e1004969. doi:10.1080/23723556.2015.1004969
  • Khurana D, Arneson LN, Schoon RA, et al. Differential regulation of human NK cell-mediated cytotoxicity by the tyrosine kinase Itk. J Immunol. 2007;178(6):3575–3582. doi:10.4049/jimmunol.178.6.3575
  • McDonald C, Xanthopoulos C, Kostareli E. The role of Bruton’s tyrosine kinase in the immune system and disease. Immunol. 2021;164(4):722–736. doi:10.1111/imm.13416
  • Kozaki R, Yasuhiro T, Kato H, et al. Investigation of the anti-tumor mechanism of tirabrutinib, a highly selective Bruton’s tyrosine kinase inhibitor, by phosphoproteomics and transcriptomics. PLoS One. 2023;18(3):e0282166. doi:10.1371/journal.pone.0282166
  • Stephens DM, Byrd JC. How I manage ibrutinib intolerance and complications in patients with chronic lymphocytic leukemia. Blood. 2019;133(12):1298–1307. doi:10.1182/blood-2018-11-846808
  • Coutre SE, Byrd JC, Hillmen P, et al. Long-term safety of single-agent ibrutinib in patients with chronic lymphocytic leukemia in 3 pivotal studies. Blood Adv. 2019;3(12):1799–1807. doi:10.1182/bloodadvances.2018028761
  • Dickerson T, Wiczer T, Waller A, et al. Hypertension and incident cardiovascular events following ibrutinib initiation. Blood. 2019;134(22):1919–1928. doi:10.1182/blood.2019000840
  • Kumar PS, Wiczer T, Rosen L, et al. Evaluation of the incidence and risk factors associated with bleeding events in patients receiving acalabrutinib therapy. Blood. 2021;138(Supplement 1):3729–3729. doi:10.1182/blood-2021-148719
  • Brown JR, Byrd JC, Ghia P, et al. Cardiovascular adverse events in patients with chronic lymphocytic leukemia receiving acalabrutinib monotherapy: pooled analysis of 762 patients. Haematologica. 2022;107(6):1335–1346. doi:10.3324/haematol.2021.278901
  • Tam CS, Dimopoulos M, Garcia-Sanz R, et al. Pooled safety analysis of zanubrutinib monotherapy in patients with B-cell malignancies. Blood Adv. 2022;6(4):1296–1308. doi:10.1182/bloodadvances.2021005621
  • Sekiguchi N, Rai S, Munakata W, et al. Two-year outcomes of tirabrutinib monotherapy in Waldenström’s macroglobulinemia. Cancer Sci. 2022;113(6):2085–2096. doi:10.1111/cas.15344
  • Xiao L, Salem J-E, Clauss S, et al. Ibrutinib-mediated atrial fibrillation attributable to inhibition of C-terminal Src kinase. Circulation. 2020;142(25):2443–2455. doi:10.1161/CIRCULATIONAHA.120.049210
  • Hillmen P, Eichhorst B, Brown JR, et al. Zanubrutinib versus ibrutinib in relapsed/refractory chronic lymphocytic leukemia and small lymphocytic lymphoma: interim analysis of a randomized phase III trial. J Clin Oncol. 2023;41(5):1035–1045. doi:10.1200/JCO.22.00510
  • O’Brien SM, Brown JR, Byrd JC, et al. Monitoring and managing BTK inhibitor treatment-related adverse events in clinical practice. Front Oncol. 2021;11:720704. doi:10.3389/fonc.2021.720704
  • Barrientos JC, O’Brien S, Brown JR, et al. Improvement in parameters of hematologic and immunologic function and patient well-being in the phase III RESONATE study of ibrutinib versus ofatumumab in patients with previously treated chronic lymphocytic leukemia/small lymphocytic lymphoma. Clin Lymphoma Myeloma Leuk. 2018;18(12):803–813.e7. doi:10.1016/j.clml.2018.08.007
  • Sibaud V, Beylot-Barry M, Protin C, et al. Dermatological toxicities of Bruton’s tyrosine kinase inhibitors. Am J Clin Dermatol. 2020;21(6):799–812. doi:10.1007/s40257-020-00535-x
  • Narita Y, Nagane M, Mishima K, et al. Phase I/II study of tirabrutinib, a second-generation Bruton’s tyrosine kinase inhibitor, in relapsed/refractory primary central nervous system lymphoma. Neuro Oncol. 2021;23(1):122–133. doi:10.1093/neuonc/noaa145
  • Munir T, Brown JR, O’Brien S, 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(12):1353–1363. doi:10.1002/ajh.25638
  • Wang ML, Blum KA, Martin P, et al. Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results. Blood. 2015;126(6):739–745. doi:10.1182/blood-2015-03-635326
  • Byrd JC, Wierda WG, Schuh A, et al. Acalabrutinib monotherapy in patients with relapsed/refractory chronic lymphocytic leukemia: updated phase 2 results. Blood. 2020;135(15):1204–1213. doi:10.1182/blood.2018884940
  • Sharman JP, Egyed M, Jurczak W, et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzmab for treatment-naive chronic lymphocytic leukaemia (ELEVATE TN): a randomised, controlled, phase 3 trial. Lancet. 2020;395(10232):1278–1291. doi:10.1016/S0140-6736(20)30262-2
  • Walter HS, Jayne S, Rule SA, et al. Long-term follow-up of patients with CLL treated with the selective Bruton’s tyrosine kinase inhibitor ONO/GS-4059. Blood. 2017;129(20):2808–2810. doi:10.1182/blood-2017-02-765115
  • Rule SA, Cartron G, Fegan C, et al. Long-term follow-up of patients with mantle cell lymphoma (MCL) treated with the selective Bruton’s tyrosine kinase inhibitor tirabrutinib (GS/ONO-4059). Leukemia. 2020;34(5):1458–1461. doi:10.1038/s41375-019-0658-7
  • Aw DC-W, Tan EH, Chin TM, et al. Management of epidermal growth factor receptor tyrosine kinase inhibitor-related cutaneous and gastrointestinal toxicities. Asia Pac J Clin Oncol. 2018;14(1):23–31. doi:10.1111/ajco.12687
  • Singer S, Tan SY, Dewan AK, et al. Cutaneous eruptions from ibrutinib resembling EGFR inhibitor-induced dermatologic adverse events. J Am Acad Dermatol. 2019;88(6):1271–1281. doi:10.1016/j.jaad.2019.12.031
  • Zhang Y, Wang W, Zhao D, et al. S224: preliminary results of a phase 2 study of orelabrutinib in combination with anti-PD-1 monoclonal antibody in refractory or relapsed primary CNS lymphoma. HemaSphere. 2022;6:125–126. doi:10.1097/01.HS9.0000843788.10370.07
  • Ghez D, Calleja A, Protin C, et al. Early-onset invasive aspergillosis and other fungal infections in patients treated with ibrutinib. Blood. 2018;131(17):1955–1959. doi:10.1182/blood-2017-11-818286
  • Bernard S, Goldwirt L, Amorim S, et al. Activity of ibrutinib in mantle cell lymphoma patients with central nervous system relapse. Blood. 2015;126(14):1695–1698. doi:10.1182/blood-2015-05-647834
  • Zhang Y, Li Y, Zhuang Z, et al. Preliminary evaluation of zanubrutinib-containing regimens in DLBCL and the cerebrospinal fluid distribution of zanubrutinib: a 13-case series. Front Oncol. 2021;11:760405. doi:10.3389/fonc.2021.760405
  • de Zwart L, Snoeys J, De Jong J, et al. Ibrutinib dosing strategies based on interaction potential of CYP3A4 perpetrators using physiologically based pharmacokinetic modeling. Clin Pharmacol Ther. 2016;100(5):548–557. doi:10.1002/cpt.419
  • Tam CS, Ou YC, Trotman J, et al. Clinical pharmacology and PK/PD translation of the second-generation Bruton’s tyrosine kinase inhibitor, zanubrutinib. Expert Rev Clin Pharmacol. 2021;14(11):1329–1344. doi:10.1080/17512433.2021.1978288
  • Grommes C, Tang SS, Wolfe J, et al. Phase 1b trial of an ibrutinib-based combination therapy in recurrent/refractory CNS lymphoma. Blood. 2019;133(5):436–445. doi:10.1182/blood-2018-09-875732
  • Choi KJ, Kim Y-Y, Jang SY, et al. Synergistic effects of BTK inhibitor HM71224 and methotrexate in a collagen-induced arthritis rat model. In Vivo. 2021;35(6):3245–3251. doi:10.21873/invivo.12619
  • Soussain C, Choquet S, Blonski M, et al. Ibrutinib monotherapy for relapse or refractory primary CNS lymphoma and primary vitreoretinal lymphoma: final analysis of the phase II ‘proof-of-concept’ iLOC study by the lymphoma study association (LYSA) and the French oculo-cerebral lymphoma (LOC) network. Eur J Cancer. 2019;117:121–130. doi:10.1016/j.ejca.2019.05.024
  • NCCN Guidelines. Central Nervous System Cancers. Version 1.2023 [cited 2023 Jul 28]. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=patients&id=53.
  • Song J, Liu H, Jiao Z, et al. Zanubrutinib, lenalidomide, rituximab, temozolomide and methotrexate (RLZT ± MTX) as first-line treatment for newly diagnosed PCNSL: a prospective, open-label, multicenter clinical trial. Blood. 2022;140(Supplement 1):3739–3740. doi:10.1182/blood-2022-164677
  • Asai K, Narita Y, Nagane M, et al. Final three-year follow-up analysis of phase I/II study on tirabrutinib in patients with relapsed or refractory primary central nervous system lymphoma. J Clin Oncol. 2023;41(16_suppl):7548–7548. doi:10.1200/JCO.2023.41.16_suppl.7548
  • Kawasaki A, Matsushita M, Yoshida S, et al. Real-world evidence for the safety and effectiveness of tirabrutinib in 140 Japanese patients with recurrent or refractory primary central nervous system lymphoma: interim report of postmarketing surveillance. Blood. 2022;140(Supplement 1):6685–6686. doi:10.1182/blood-2022-156472
  • Alcantara M, Chevrier M, Jardin F, et al. Final analysis of the phase IB part of the LOC-R01 trial, a non-comparative randomized phase IB/II study of escalating doses of lenalidomide and ibrutinib in association with R-MPV for patients with a newly diagnosed primary central nervous system lymphoma (PCNSL). J Clin Oncol. 2023;41(16_suppl):7566–7566. doi:10.1200/JCO.2023.41.16_suppl.7566
  • Garcia-Reyero J, Martinez Magunacelaya N, Gonzalez Pereña A, et al. Clonal evolution in primary diffuse large B-cell lymphoma of the central nervous system. Appl Immunohistochem Mol Morphol. 2020;28(8):e68–e71. doi:10.1097/PAI.0000000000000655
  • Green K, Hogg JP. Central nervous system lymphoma. Treasure Island (FL): StatPearls Publishing; 2022. Available from: https://www.ncbi.nlm.nih.gov/books/NBK545145/
  • Simard J, Roschewski M. SOHO state of the art updates and next questions: prophylaxis and management of secondary CNS lymphoma. Clin Lymphoma Myeloma Leuk. 2022;22(10):709–717. doi:10.1016/j.clml.2022.06.002
  • Grommes C, Wolfe J, Gavrilovic I, et al. Phase II of single-agent ibrutinib in recurrent/refractory primary (PCNSL) and secondary CNS lymphoma (SCNSL). Blood. 2018;132(Supplement 1):2965–2965. doi:10.1182/blood-2018-99-118538
  • Wilson WH, Young RM, Schmitz R, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21(8):922–926. doi:10.1038/nm.3884
  • Severson EA, Haberberger J, Hemmerich A, et al. Genomic profiling reveals differences in primary central nervous system lymphoma and large B-cell lymphoma, with subtyping suggesting sensitivity to BTK inhibition. Oncologist. 2023;28(1):e26–e35. doi:10.1093/oncolo/oyac190
  • Woyach JA, Furman RR, Liu T-M, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370(24):2286–2294. doi:10.1056/NEJMoa1400029
  • Sedlarikova L, Petrackova A, Papajik T, et al. Resistance-associated mutations in chronic lymphocytic leukemia patients treated with novel agents. Front Oncol. 2020;10:894. doi:10.3389/fonc.2020.00894
  • Iskierka-Jażdżewska E, Obracaj A, Urbaniak M, et al. New treatment options for newly-diagnosed and relapsed chronic lymphocytic leukemia. Curr Treat Options Oncol. 2022;23(6):775–795. doi:10.1007/s11864-022-00974-0
  • Wierda WG, Lewis DJ, Ghia P, et al. Efficacy of pirtobrutinib, a highly selective, non-covalent (reversible) BTK inhibitor in Richter transformation: results from the phase 1/2 BRUIN study. Blood. 2022;140(Supplement 1):846–849. doi:10.1182/blood-2022-157058
  • Ran F, Liu Y, Xu Z, et al. Recent development of BTK-based dual inhibitors in the treatment of cancers. Eur J Med Chem. 2022;233:114232. doi:10.1016/j.ejmech.2022.114232