Despite the remarkable success of targeted therapies for mantle cell lymphoma (MCL), including inhibitors of Bruton tyrosine kinase (BTK) and CD19-directed chimeric antigen receptor (CAR) T-cell therapy, resistance and disease relapse persist, so there is an urgent need to develop novel agents and combinatorial strategies against this deadly disease.1,2 BTK is a key component of the B-cell receptor pathway, which regulates B-cell survival and proliferation. Ibrutinib, the first Food and Drug Administration-approved covalent BTK inhibitor, achieved overall response rates of 70-77% in patients with relapsed/refractory MCL,3 which represented a major milestone in targeted MCL therapies.4 As a key regulator of apoptosis, BCL-2 is aberrantly expressed in MCL, and its inhibition with venetoclax (ABT-199) induces massive apoptosis in MCL cells.5 Notably, combinatorial ibrutinib and venetoclax yielded favorable complete response rates in MCL patients in a phase II study (71%)6 and in the phase III SYMPATICO study (62%),3 indicating that novel combinatorial approaches can be useful to overcome therapeutic resistance and increase durability of effective treatments for MCL.
Pirtobrutinib (LOXO-305) is a next-generation, highly selective, non-covalent BTK inhibitor.7 Compared to traditional covalent BTK inhibitors, pirtobrutinib achieves remarkable target coverage regardless of the intrinsically high rate of BTK turnover, and lacks the off-target inhibition of other kinases.8 In the phase I/II BRUIN study pirtobrutinib exhibited promising efficacy in heavily pretreated MCL patients irrespective of prior exposure to covalent BTK inhibitors.9 Given the clinical success of combinatorial ibrutinib and venetoclax in MCL patients, we investigated and here report the antitumor effects of pirtobrutinib in combination with venetoclax in various MCL models in vitro and in vivo to provide proof of concept for further exploration in the clinic. The patients’ apheresis samples used in this study were collected after obtaining informed consent and approval from the Institutional Review Board at The University of Texas MD Anderson Cancer Center and all experimental procedures and protocols were approved by the Institutional Animal Care and Use Committee of The University of Texas MD Anderson Cancer Center.
First, we performed in vitro cell viability assays to test the efficacy of the combination of pirtobrutinib and venetoclax in a panel of MCL cell lines and primary MCL patients’ samples (Figure 1A, B). Compared to the effects of single agents, the cytotoxicity of the combination was enhanced in all the cells tested irrespective of their response to ibrutinib, venetoclax, and even anti-CD19 CAR T-cell therapy, indicating that this novel combination has promising potential for overcoming multiple types of therapeutic resistance in MCL. A dose-response viability assay for BTK knockdown JeKo-1 cells (JeKo BTK KD_2) (Online Supplementary Figure S1A)10 and ibrutinib-resistant JeKo-1 cells (JeKo-ibrutinib-R)11 showed a clear synergistic effect in reducing cell viability with combination indexes12 <1 (0.54 and 0.47, respectively) (Online Supplementary Figure S1B). As a functional outcome, the annexin V/propidium iodide apoptosis assay revealed enhanced cytotoxicity of the combination compared to single agents in ibrutinib-resistant cell lines (JeKo BTK KD cells and JeKo-ibrutinib-R) and a venetoclax-resistant cell line (Mino-venetoclax-R)13 (Online Supplementary Figure S1C). Consistent with this, reverse-phase protein array analysis on JeKo-ibrutinib-R cells confirmed enhanced apoptosis for the combination, as demonstrated by increases in the stress response proteins p-NDRG1 and p-JNK, the DNA damage marker γH2AX, and cleaved apoptotic caspases (Online Supplementary Figure S1D). In further accordance with these results, western blotting assay verified that the late-stage cell death markers cleaved caspase 3 and cleaved PARP were markedly increased by the combinatorial treatment (Online Supplementary Figure S1E).
To determine the in vivo anti-MCL efficacy of pirtobrutinib and venetoclax, we tested the combination in an aggressive patient-derived xenograft mouse model generated from a dual ibrutinib- and CD19-targeted CAR T-cell-resistant patient (PT15 in Figure 1B). Compared to a vehicle, each of the single-drug treatments decreased tumor growth in mice, as confirmed by measurement of both tumor volume (Figure 1C) and levels of the tumor marker b2-microglobulin in mouse serum (Online Supplementary Figure S2A). Strikingly, the combination therapy completely suppressed tumor development long after the mice in the other three groups had reached the humane endpoint, and caused no noticeable adverse effects in the mice (e.g., loss of body weight or hair) during the entire treatment (Online Supplementary Figure S2B). The combination treatment was discontinued on day 85 to track the effect on tumor progression. The mice remained tumor-free for more than 2 months. Kaplan-Meier survival analysis demonstrated that the combination therapy dramatically prolonged mouse survival with a median survival longer than 150 days, compared to 81 days for the mice treated with pirtobrutinib and 80 days for those given venetoclax, indicating that this novel combination regimen may be a promising strategy to overcome ibrutinib- and CAR T-cell therapy dual-resistant MCL (Figure 1D).
To validate this in vivo finding, we generated a xenograft model by inoculating Mino-venetoclax-R cells subcutaneously into NSG mice. The tumor-bearing mice were administered pirtobrutinib and venetoclax alone or in combination for 25 days and then euthanized. The monotherapy moderately reduced the rate of tumor growth, while the combinatorial treatment almost completely prevented tumor growth, as reflected by tumor volume and mass (Figure 2A, B). These results indicate that the combination holds promise to overcome venetoclax resistance in MCL. To evaluate the transcriptome determinants associated with this combination, whole-transcriptome RNA sequencing studies were performed on tumor cells harvested from mice treated with vehicle, pirtobrutinib, venetoclax, and the combination of the latter two. A total of 967 genes were differentially expressed in the combined treatment group relative to the group given the vehicle control, of which 478 genes were significantly upregulated while 489 genes were downregulated (absolute [log fold change] >1 and adjusted P value <0.05) (Figure 2C). In terms of the genes that were differentially expressed in cells from animals exposed to the combination therapy but not to either monotherapy relative to vehicle, 224 genes (51.9%) were found to be upregulated and 251 genes (60.8%) were downregulated (adjusted P value <0.05) (Figure 2D). Gene set enrichment analysis was performed to determine the associated cancer hallmark gene sets. Compared to single treatments alone and vehicle, combination therapy suppressed mTORC1 signaling, MYC targets, E2F targets, oxidative phosphorylation (OXPHOS), fatty acid metabolism, and adipogenesis pathways (false discovery rate <0.1) (Figure 2E, Online Supplementary Figure S3). It is noteworthy that upregulation of these oncogenic and metabolic pathways has been previously reported to drive ibrutinib resistance in MCL.14,15 Furthermore, not only OXPHOS but also glycolysis, the major energy production pathway, was downregulated in the combination group, indicating that mitochondrial energy production for cancer cells to grow and survive was efficiently blocked by this combination therapy.
In summary, our findings demonstrated that the combination of pirtobrutinib and venetoclax had enhanced antitumor efficacy over both monotherapies in preclinical, resistant MCL models and support future investigation of this promising regimen in other B-cell malignancies. Transcriptome profiling revealed a significantly downregulated gene expression signature associated with oncogenic MYC targets, mTORC1 signaling, and metabolic pathways such as glycolysis and OXPHOS. Further mechanistic studies are warranted to elucidate the underpinnings of this combinatorial efficacy. A phase II clinical trial (NCT05529069) based on this study has been activated at MD Anderson Cancer Center.
Footnotes
- Received September 1, 2022
- Accepted November 28, 2022
Correspondence
Disclosures
MW is a consultant to AbbVie, Acerta Pharma, AstraZeneca, BeiGene, BioInvent, Deciphera, InnoCare, Janssen, Kite Pharma, Leukemia & Lymphoma Society, Lilly, Merck, Milken Institute, Oncternal, Parexel, Pepromene Bio, Pharmacyclics, and VelosBio, and he has received research support from Acerta Pharma, AstraZeneca, BeiGene, BioInvent, Celgene, Genmab, Genentech, Innocare, Janssen, Juno Therapeutics, Kite Pharma, Lilly, Loxo Oncology, Molecular Templates, Oncternal, Pharmacyclics, VelosBio and Vincerx. MW has also received speaker’s honoraria from AbbVie, Acerta Pharma, AstraZeneca, BeiGene, BioInvent, Dava Oncology, Eastern Virginia Medical School, IDEOlogy Health, Janssen, Kite Pharma, Leukemia & Lymphoma Society, LLC TS Oncology, Medscape, Meeting Minds Experts, MJH Life Sciences, Merck, Moffit Cancer Center, Oncology Specialty Group, OncLive, Pharmacyclics, Physicians Education Resources (PER), Practice Point Communications (PPC), and Studio ER Congressi. All other authors declare that they have no competing financial interests.
Contributions
MW and YL conceived and designed the study; YL, VCJ, AJ, JM, YL, YC, IH and WW performed the experiments; FY and YL analyzed the data; YL wrote the manuscript; MW, FY, HL, YY, LN and JJ edited the manuscript.
Data-sharing statement
The original data and protocols can be obtained upon reasonable request.
Funding
Acknowledgments
The authors thank the patients and their families for their contribution to this research study. Thanks to Paul C. Dolber and Numsen Hail for offering help in reviewing and revising the manuscript.
References
- Wang M, Munoz J, Goy A. KTE-X19 CAR T-cell therapy in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2020; 382(14):1331-1342. https://doi.org/10.1056/NEJMoa1914347PubMedPubMed CentralGoogle Scholar
- Jain P, Nastoupil L, Westin J. Outcomes and management of patients with mantle cell lymphoma after progression on brexucabtagene autoleucel therapy. Br J Haematol. 2021; 192(2):e38-e42. https://doi.org/10.1111/bjh.17197PubMedGoogle Scholar
- Wang M, Ramchandren R, Chen R. Concurrent ibrutinib plus venetoclax in relapsed/refractory mantle cell lymphoma: the safety run-in of the phase 3 SYMPATICO study. J Hematol Oncol. 2021; 14(1):179. https://doi.org/10.1186/s13045-021-01188-xPubMedPubMed CentralGoogle Scholar
- Wen T, Wang J, Shi Y, Qian H, Liu P. Inhibitors targeting Bruton's tyrosine kinase in cancers: drug development advances. Leukemia. 2021; 35(2):312-332. https://doi.org/10.1038/s41375-020-01072-6PubMedPubMed CentralGoogle Scholar
- Kapoor I, Bodo J, Hill BT, Hsi ED, Almasan A. Targeting BCL-2 in B-cell malignancies and overcoming therapeutic resistance. Cell Death Dis. 2020; 11(11):941. https://doi.org/10.1038/s41419-020-03144-yPubMedPubMed CentralGoogle Scholar
- Tam CS, Anderson MA, Pott C. Ibrutinib plus venetoclax for the treatment of mantle-cell lymphoma. N Engl J Med. 2018; 378(13):1211-1223. https://doi.org/10.1056/NEJMoa1715519PubMedGoogle Scholar
- Liu Y, Jiang C, Yan F. Pirtobrutinib overcomes ibrutinib and venetoclax resistance in mantle cell lymphoma. Blood. 2021; 138(Supplement 1):1182-1182. https://doi.org/10.1182/blood-2021-151401Google Scholar
- 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
- 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
- Li CJ, Jiang C, Liu Y. Pleiotropic action of novel Bruton's tyrosine kinase inhibitor BGB-3111 in mantle cell lymphoma. Mol Cancer Ther. 2019; 18(2):267-277. https://doi.org/10.1158/1535-7163.MCT-18-0478PubMedPubMed CentralGoogle Scholar
- Wang J, Wang J, Lopez E. Repurposing auranofin to treat TP53-mutated or PTEN-deleted refractory B-cell lymphoma. Blood Cancer J. 2019; 9(12):95. https://doi.org/10.1038/s41408-019-0259-8PubMedPubMed CentralGoogle Scholar
- Chou T-C. The combination index (CI < 1) as the definition of synergism and of synergy claims. Synergy. 2018; 7:49-50. https://doi.org/10.1016/j.synres.2018.04.001Google Scholar
- Pham LV, Huang S, Zhang H. Strategic therapeutic targeting to overcome venetoclax resistance in aggressive B-cell lymphomas. Clin Cancer Res. 2018; 24(16):3967-3980. https://doi.org/10.1158/1078-0432.CCR-17-3004PubMedGoogle Scholar
- Zhang L, Yao Y, Zhang S. Metabolic reprogramming toward oxidative phosphorylation identifies a therapeutic target for mantle cell lymphoma. Sci Transl Med. 2019; 11(491):eaau1167. https://doi.org/10.1126/scitranslmed.aau1167PubMedGoogle Scholar
- Zhao X, Wang MY, Jiang H. Transcriptional programming drives ibrutinib-resistance evolution in mantle cell lymphoma. Cell Rep. 2021; 34(11):108870. https://doi.org/10.1016/j.celrep.2021.108870PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.