Several Bruton’s tyrosine kinase inhibitors (BTKi) have been approved for the treatment of B-cell malignancies and are particularly active in chronic lymphocytic leukemia (CLL), where they have transformed the treatment paradigm. However, the activity of currently available BTKi (ibrutinib, acalabrutinib, and zanubrutinib) requires covalent bond formation with cysteine 481 (C481) of BTK; hence resistance to covalent BTKi may be mediated through mutations which remove C481. In order to overcome this resistance, and potentially prevent the proliferation of C481 mutant cells, non-covalent BTKi that do not require interaction with the C481 residue were developed.1 This phase Ib/II trial investigated the safety and clinical activity of vecabrutinib, a reversible, non-covalent inhibitor of BTK that inhibits both wild-type and C481-mutated BTK,2 in patients with advanced, BTKiresistant B-cell malignancies. The results of the completed phase Ib dose-escalation portion of the study demonstrated that vecabrutinib was well-tolerated up to 410 mg twice daily (BID), the highest dose studied. Evidence of clinical benefit was observed, with a best response of partial response (PR) in one CLL patient and stable disease (SD) in 13 patients. Pharmacokinetics (PK) were approximately dose-proportional and sustained reductions in serum cytokine concentrations were observed at higher dose levels, suggesting BTK inhibition. However, the association between vecabrutinib dose, pharmacodynamics (PD), and clinical activity was inconsistent and the activity observed was considered insufficient for phase II expansion in patients with BTKi-resistant CLL.
Patient and disease characteristics are summarized in the Online Supplementary Table S1. Thirty-nine patients with histologically confirmed, relapsed/refractory CLL or other B-cell malignancies and ≥2 prior lines of standard systemic therapy, including progression during BTKi therapy, were enrolled and treated across seven dose-levels. The majority (77%) of patients had CLL. The enrolled population was high-risk, with a median of four prior therapies (range, 2-9), 17p deletion or TP53 mutation in 74% of all patients, and BTK C481 mutations in 55% of CLL patients.
Vecabrutinib capsules were administered orally BID at a starting dose of 20.5 mg/dose (41 mg total daily dose). Vecabrutinib was well-tolerated up to 410 mg BID (820 mg total daily dose), the highest dose studied. One patient, treated at the 41 mg BID dose level, experienced a doselimiting toxicity (DLT), consisting of failure to receive >80% of planned vecabrutinib doses due to adverse events (AE) of grade 3 alanine aminotransferase (ALT) and grade 2 aspartate aminotransferase (AST) elevations. Upon expansion of this cohort, no additional DLT were observed and dose escalation continued. The maximum-tolerated dose of vecabrutinib was not reached.
The most common treatment-emergent AE were anemia (31%) and nausea, fatigue, headache and dyspnea (21% each). The most common AE considered treatmentrelated by the investigator were anemia and fatigue (10% each). Grade ≥3 AE were mainly hematologic, including anemia (23%), neutropenia (13%) and thrombocytopenia (10%) (Online Supplementary Table S2). Grade ≥3 AE considered treatment-related by the investigator consisted of leukocytosis in two patients (5.1%) and anemia, neutropenia, and increased AST in one patient (2.6%) each. No obvious pattern of dose-dependent toxicity was observed, with no grade ≥3 AE observed at the two highest dose levels. One or more serious AE (SAE) were reported in seven patients and consisted of cellulitis (in 2 patients) and lymphocytosis, intestinal perforation, myelitis, sepsis, hematuria, pleural effusion, and deep vein thrombosis (in 1 patient each). No SAE were considered related to study treatment per the investigator. Two patient deaths were associated with AE: perforated bowel in one patient treated at the 41 mg BID dose level (who had mantle cell lymphoma [MCL] with bowel involvement), and sepsis in one patient treated at the 164 mg BID dose level. Neither event was considered related to study treatment. There were no cardiac events or clinically significant electrocardiogram findings.
Vecabrutinib showed modest evidence of clinical benefit, with one PR observed in a patient with CLL (treated at the 246 mg BID dose level) and SD in 13 patients (31%; 11 CLL, 1 MCL, 1 marginal zone lymphoma). A waterfall plot of percent change in tumor burden from baseline in all patients is displayed in Figure 1. Among 14 patients with a best response of PR or SD, six had BTK C481 mutations, eight had 17p deletion and/or TP53 mutations, and the median number of prior therapies was three (range, 2-9). Patients with PR or SD remained on study treatment for a median of 28.5 weeks (range, 6.1-56+ weeks). Eight of these patients received vecabrutinib for ≥6 months, including five who discontinued treatment due to termination of the study and not for progressive disease, suggesting some durable clinical benefit.
Pharmacokinetic data were available for 38 patients. Concentration-time profiles and linear regression analysis indicated that both exposure and median steady-state Ctrough concentrations generally increased in an approximately dose-proportional manner. Exposure to vecabrutinib was maintained across the ~12 hour dosing interval, supporting BID dosing, with cycle 1 day 8 trough values at dose levels ≥164 mg BID expected to provide >90% inhibition of BTK signaling based an earlier single-dose phase I study.3
Pharmacodynamic activity of vecabrutinib was assessed in CLL patients who completed cycle 1 (n=25) via measurement of serum cytokine levels. CCL3, CCL4, and TNFa************have previously been shown to be inhibited by other BTKi.4–6 For all three cytokines, a sustained reduction in serum concentration was evident in most patients after one cycle of vecabrutinib treatment at higher dose levels (246, 328 and 410 mg), suggesting inhibition of BTK activity. Mean reductions at these dose levels ranged from 34-62% for CCL3, 33-59% for CCL4, and 24-57% for TNFa************(Figure 2). In regression analyses, there was a trend for greater reduction in serum cytokine levels with increasing Cmax and AUClast as well as vecabrutinib dose, suggestive of an exposure-response effect (Online Supplementary Figure S1). Chemokine reduction was associated with clinical benefit, with decreased serum cytokine levels demonstrated in all but one patient with a clinical response of PR or SD. However, the extent of inhibition was generally less than that observed in BTKi-naïve patients treated with ibrutinib (which produced median decreases ≥80% for CCL3, CCL4 and TNFa),6 consistent with the limited clinical activity observed for vecabrutinib.
The question arises as to why BTK inhibition by vecabrutinib did not translate to a clinical response despite strong preclinical evidence and promising early (phase Ia) clinical PK/PD data in healthy subjects, particularly in CLL patients in whom substantial clinical activity has been observed with other non-covalent BTKi. In vitro cellular assays were performed to evaluate the half maximal inhibitory concentration (IC50) values and BTK residence time (i.e., the time a BTKi remains bound to BTK) for vecabrutinib compared with other BTKi to look for potential correlation with outcome (Table 1). IC50 values for vecabrutinib (18.4 nM) and ARQ 531 (32.9 nM) were similar, however, ARQ 531 demonstrated greater clinical efficacy; conversely, fenebrutinib demonstrated greater in vitro potency (7.04 nM) but showed limited clinical activity in patients previously treated with ibrutinib.7–10 The residence time observed for vecabrutinib (15 minutes [min]) was much shorter than that observed for ARQ 531 (128 min) and fenebrutinib (557 min) and was also shorter relative to reported values for pirtobrutinib (LOXO-305; 314 min). Other possible explanations for the limited clinical activity observed are that vecabrutinib is highly protein bound (98.7%), which may have affected the availability of the free drug, or that vecabrutinib may not have been consistently distributed from blood to disease sites; either of these possibilities may have resulted in levels insufficient to provide adequate BTK inhibition. Furthermore, PK properties differ among non-covalent BTKi: the effective agents, pirtobrutinib and ARQ 531, have longer half-lives (approximately 20 hours and 55 hours, respectively) than the agents with limited clinical activity, vecabrutinib, fenebrutinib and dasatinib (reported half-lives ranging between 4 and 14 hours).3,7,9–12 Although no single property aligned consistently with the observed clinical activity, these attributes provide possible explanations regarding the limited clinical activity observed with vecabrutinib compared to other reversible BTK inhibitors.
Overall, vecabrutinib was well-tolerated and demonstrated some evidence of clinical benefit. However, despite dose-proportional PK, the association between vecabrutinib dose, PD, and clinical response was inconsistent. Increasing the dose from 246 to 410 mg BID did not uniformly correlate with increased PD activity though there was an overall trend towards improved inhibition with dose. Assessment of clinical activity by dose may have been confounded by the impact of baseline patient characteristics: clinical benefit (i.e., PR or SD lasting >6 months) was most commonly observed in patients who were less heavily pretreated and had better prognostic factors as identified by Ahn et al.,13 such as lower baseline lactate dehydrogenase levels and wild-type TP53, regardless of dose. These results suggest that the potency of singleagent vecabrutinib was not sufficient to control disease in refractory patients; however vecabrutinib in combination with other agents, including BCL2 inhibitors,14 may result in improved efficacy. Based on the dose-escalation results, the activity observed in BTKi-resistant patients at the dose levels studied was considered insufficient for phase II expansion of this patient cohort. Future directions for vecabrutinib may include indications such as chronic graftversus- host disease or in combination with chimeric antigen receptor T-cell therapies, where dual inhibition of BTK and IL-2 Inducible T-cell Kinase (ITK) may contribute to clinical outcomes.
Footnotes
- Received September 24, 2021
- Accepted December 7, 2021
Correspondence
Disclosures: JNA served as a consultant to AbbVie, Acerta, Ascentage Pharma, AstraZeneca, Bayer, BeiGene, Epizyme, Genentech, Janssen, Pharmacyclics, Sunesis Pharmaceuticals, TG Therapeutics, and Verastem Oncology; received honoraria from AbbVie, BeiGene, Janssen, Pharmacyclics; and received research funding from AstraZeneca, Celgene, Genentech, and Janssen. JP-I served as a consultant to AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Takeda, Teva, and TG Therapeutics; and served on the speakers bureau for AbbVie, Bayer, Janssen, Sanofi, and Takeda. DEG reports no conflict of interest. KP served as a consultant to AstraZeneca, Celgene, Genentech, Pharmacyclics/Janssen and Sunesis; received research funding from AstraZeneca; and was a member of the Speakers Bureau for AstraZeneca, Celgene, Genentech, and Pharmacyclics/Janssen. JPS served as a consultant to and received honoraria and research funding from AbbVie, Acerta, AstraZeneca, Genentech, Janssen, Pharmacyclics, and TG Therapeutics. WGW received research funding from AbbVie, Acerta, Cyclcel, Genentech, Gilead Sciences, GSK/Novartis’ Janssen, Juno Therapeutics, KITE pharma, Loxo Oncology, Miragen, Oncternal Therapeutics, Pharmacyclics, Sunesis, and Xencor. MYC served as a consultant to AbbVie, Genentech, Gilead, Pharmacyclics, and Rigel; received research funding from AbbVie, Oncternal Therapeutics, Pharmacyclics, and Rigel; and was a member of the Speakers Bureau for AbbVie, Genentech, Gilead, and Pharmacyclics. SMO served as a consultant to AbbVie, Alexion, Amgen, Aptose Biosciences, Astellas, Celgene, Eisai, Gilead, GlaxoSmithKline, Janssen, Pfizer, Pharmacyclics, Sunesis, TG Therapeutics, Vaniam Group, and Verastem Oncology; received honoraria from AbbVie, Janssen, and Pfizer; and received research funding from Acerta, Gilead, KITE pharma, Pfizer, Pharmacyclics, Regeneron, Sunesis, and TG Therapeutics. MS served as a consultant to AbbVie, ADC Therapeutics, AstraZeneca, Atara Biotherapeutics, Genentech, Gilead, Pharmacyclics, Sound Biologics, and Verastem; and received research funding from AbbVie, Acerta Pharma, BeiGene, Celgene, Genentech, Gilead, Mustang Bio, Pharmacyclics, Sunesis, TG Therapeutics, and Verastem. MSD served as a consultant to AbbVie, Adaptive Biotechnologies, Ascentage Pharma, AstraZeneca, BeiGene, BMS, Celgene, Eli Lilly, Genentech, Janssen, MEI Pharma, Merck, Novartis, Takeda, and Verastem; received research funding from Ascentage Pharma, AstraZeneca, BMS, Genentech, MEI pharma, Novartis, Pharmacyclics, Surface Oncology, TG Therapeutics, and Verastem; and received honoraria from Research to Practice. JMP served as a consultant to AstraZeneca, Gilead Sciences, and Pharmacyclics. HAY was a member of the Speakers Bureau for Amgen, AstraZeneca, BeiGene, Janssen, Karyopharm, Pharmacylics, and Sanofi; and holds publicly-traded stock in Karyopharm. RW served as a consultant to Sunesis Pharmaceuticals. GA served as a consultant to Sunesis Pharmaceuticals. PT was employed by Sunesis Pharmaceuticals. JAF was employed by Sunesis Pharmaceuticals. DLC served as a consultant to Sunesis Pharmaceuticals. RRF served as a consultant to AbbVie, Acerta Pharma, AstraZeneca, BeiGene, Genentech, Incyte, Janssen, Loxo Oncology, Morphosys, Oncotracker, Pharmacyclics, Sanofi, Sunesis, TG Therapeutics, and Verastem. JRB served as a consultant to AbbVie, Acerta Pharma, AstraZeneca, BeiGene, Catapult Therapeutics, Dynamo, Genentech, Gilead, Juno/Celgene, Kite, Loxo, Novartis, Octapharma, Pfizer, Pharmacyclics, Sunesis, TG Therapeutics, and Verastem; received honoraria from Janssen and Teva, received research funding from Gilead, Kite, Loxo, Sun Pharmaceuticals, and Verastem; and was a member of the data safetymonitoring board for Ivectys and Morphosys.
Contributions: JNA, JP-I, DEG, KP, JPS, WGW, MYC, SMO, MS, MSD, JMP, HAY, RW, GA, PT, JAF, RRF, and JRB contributed to study concept and study supervision; PT and JAF were responsible for study administration; JNA, JP-I, DEG, KP, JPS, WGW, MYC, SMO, MS, MSD, JMP, HAY, RRF, and JRB provided patients and conducted the investigation; DC did a formal analysis of the data; DC, PT, and JAF prepared the data presentation; JA, GA, PT, JAF, and JRB wrote the original draft of the manuscript. All authors reviewed and edited the manuscript.
Trial registration: clinicaltrials.gov Identifier: NCT03037645; EU clinical trials register identifier: EudraCT # 2018-000108-41
Funding
financial support for this study was provided by Sunesis Pharmaceuticals, Inc., South San Francisco, CA.
Acknowledgments
Medical writing support was provided by Janis Leonoudakis, PhD, and was funded by Sunesis Pharmaceuticals, Inc., San Francisco, CA.
References
- Gu D, Tang H, Wu J, Li J, Miao Y. Targeting Bruton tyrosine kinase using non-covalent inhibitors in B cell malignancies. J Hematol Oncol. 2021; 14(1):40. https://doi.org/10.1186/s13045-021-01049-7PubMedPubMed CentralGoogle Scholar
- Allan JA, Patel K, Mato AR. Preliminary results of a phase 1b/2 dose-escalation and cohort-expansion study of the noncovalent, reversible Bruton’s tyrosine kinase inhibitor (BTKi), vecabrutinib, in B-cell malignancies. HemaSphere. 2019; 3(suppl 1):520. https://doi.org/10.1097/01.HS9.0000562876.57990.65Google Scholar
- Neuman L, Ward R, Arnold D. First-in-human phase 1a study of the safety, pharmacokinetics, and pharmacodynamics of the noncovalent bruton tyrosine kinase (BTK) inhibitor SNS-062 in healthy subjects. Blood. 2016; 128(Suppl):S2032. https://doi.org/10.1182/blood.V128.22.2032.2032Google Scholar
- Ponader S, Chen S-S, Buggy JJ. The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood. 2012; 119(5):1182-1189. https://doi.org/10.1182/blood-2011-10-386417PubMedPubMed CentralGoogle Scholar
- Byrd JC, Harrington B, O’Brien S. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016; 374(4):323-332. https://doi.org/10.1056/NEJMoa1509981PubMedPubMed CentralGoogle Scholar
- Niemann CU, Herman SEM, Maric I. Disruption of in vivo chronic lymphocytic leukemia tumor-microenvironment interactions by ibrutinib - findings from an investigator-initiated phase II study. Clin Cancer Res. 2016; 22(7):1572-1582. https://doi.org/10.1158/1078-0432.CCR-15-1965PubMedPubMed CentralGoogle Scholar
- Byrd JC, Smith S, Wagner-Johnston N. First-in-human phase 1 study of the BTK inhibitor GDC-0853 in relapsed or refractory Bcell NHL and CLL. Oncotarget. 2018; 9(16):13023-13035. https://doi.org/10.18632/oncotarget.24310PubMedPubMed CentralGoogle Scholar
- Crawford JJ, Johnson AR, Misner DL. Discovery of GDC-0853: a potent, selective, and noncovalent Bruton’s tyrosine kinase inhibitor in early clinical development. J Med Chem. 2018; 61(6):2227-2245. https://doi.org/10.1021/acs.jmedchem.7b01712PubMedGoogle Scholar
- Herman AE, Chinn LW, Kotwal SG. Safety, pharmacokinetics, and pharmacodynamics in healthy volunteers treated with GDC- 0853, a selective reversible Bruton’s tyrosine kinase inhibitor. Clin Pharmacol Ther. 2018; 103(6):1020-1028. https://doi.org/10.1002/cpt.1056PubMedGoogle Scholar
- Woyach J, Stephens DM, Flinn IW. Final results of phase 1, dose escalation study evaluating ARQ 531 in patients with relapsed or refractory B-cell lymphoid malignancies. Blood. 2019; 134(Suppl 1):S4298. https://doi.org/10.1182/blood-2019-127260Google Scholar
- Christopher LJ, Cui D, Wu C. Metabolism and disposition of dasatinib after oral administration to humans. Drug Metab Dispos. 2008; 36(7):1357-1364. https://doi.org/10.1124/dmd.107.018267PubMedGoogle Scholar
- Mato AR, Shah NN, Jurczak W. Pirtobrutinib in relapsed or refractory B-cell malignancies (BRUIN): a phase 1/2 study. Lancet. 2021; 397(10277):892-901. https://doi.org/10.1016/S0140-6736(21)00224-5PubMedGoogle Scholar
- Ahn IE, Tian X, Ipe D. Prediction of outcome in patients with chronic lymphocytic leukemia treated with ibrutinib: development and validation of a four-factor prognostic model. J Clin Oncol. 2021; 39(6):576-585. https://doi.org/10.1200/JCO.20.00979PubMedPubMed CentralGoogle Scholar
- Jebaraj B, Müller A, Dheenadayalan R. Evaluation of vecabrutinib as a model for non-covalent BTK/ITK inhibition for treatment of chronic lymphocytic leukemia. Blood. 2022; 139(6):859-875. https://doi.org/10.1182/blood.2021011516PubMedGoogle Scholar
- Gomez EB, Isabel L, Rosendahal MS, Rothenberg SM, Andrews SW, Brandhuber BJ. Loxo-305, a highly selective and non-covalent next generation BTK inhibitor, inhibits diverse BTK C481 substitution mutations. Blood. 2019; 135(Suppl 1):S4644. https://doi.org/10.1182/blood-2019-126114Google Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.