Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2667 2023-05-18 15:59:29 |
2 format Meta information modification 2667 2023-05-19 07:33:03 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Eyre, T.; Riches, J. Therapies Targeting Bruton Tyrosine Kinase for CLL. Encyclopedia. Available online: https://encyclopedia.pub/entry/44518 (accessed on 17 August 2024).
Eyre T, Riches J. Therapies Targeting Bruton Tyrosine Kinase for CLL. Encyclopedia. Available at: https://encyclopedia.pub/entry/44518. Accessed August 17, 2024.
Eyre, Toby, John Riches. "Therapies Targeting Bruton Tyrosine Kinase for CLL" Encyclopedia, https://encyclopedia.pub/entry/44518 (accessed August 17, 2024).
Eyre, T., & Riches, J. (2023, May 18). Therapies Targeting Bruton Tyrosine Kinase for CLL. In Encyclopedia. https://encyclopedia.pub/entry/44518
Eyre, Toby and John Riches. "Therapies Targeting Bruton Tyrosine Kinase for CLL." Encyclopedia. Web. 18 May, 2023.
Therapies Targeting Bruton Tyrosine Kinase for CLL
Edit

The development of inhibitors of Bruton tyrosine kinase (BTK) and B-cell lymphoma 2 (BCL2) has resulted in a paradigm shift in the treatment of chronic lymphocytic leukaemia (CLL). Observations regarding the importance of B-cell receptor signalling for the survival and proliferation of CLL cells led to the development of the first-in-class BTK inhibitor (BTKi), ibrutinib, for the treatment of CLL. Despite being better tolerated than chemoimmunotherapy, ibrutinib does have side effects, some of which are due to the off-target inhibition of kinases other than BTK. As a result, more specific inhibitors of BTK were developed, such as acalabrutinib and zanubrutinib, which have demonstrated equivalent/enhanced efficacy and improved tolerability in large randomized clinical trials. Despite the increased specificity for BTK, side effects and treatment resistance remain therapeutic challenges. As these drugs all bind covalently to BTK, an alternative approach was to develop noncovalent inhibitors of BTK, including pirtobrutinib and nemtabrutinib. The alternative mechanisms of BTK-binding of these agents has the potential to overcome resistance mutations, something that has been borne out in early clinical trial data.

chronic lymphocytic leukaemia Bruton tyrosine kinase ibrutinib acalabrutinib zanubrutinib

1. Ibrutinib: The “First in Class” BTKi

Ibrutinib, initially known as PCI-32765, was the first BTKi to enter clinical testing. Ibrutinib is an orally bioavailable small molecule which binds irreversibly to a cysteine residue (C481) in the BTK kinase domain to inhibit its enzymatic activity [1]. BTK could already be inferred as a critical component of BCR signalling and therefore humoral immunity due the observations made by Bruton regarding X-linked agammaglobulinemia (XLA) [2][3][4]. Individuals with XLA have mutations in the BTK gene (present on the long arm of the X-chromosome) and an immunodeficiency characterized by the absence of mature B-cells, resulting in severe antibody deficiency and recurrent infections that can manifest as soon as the protective effect of maternal immunoglobulins wanes at around 3–6 months of age. Preclinical observations confirmed the importance of BTK in CLL, demonstrating overexpression of this kinase in the leukemic cells compared with healthy B-cells, with ibrutinib demonstrating preferential, albeit modest, killing of CLL cells in vitro [5].
An initial phase 1b/2 study in 85 patients with relapsed/refractory CLL or small lymphocytic lymphoma (SLL) compared two different doses of ibrutinib, 420 mg versus 840 mg [6]. Interestingly, the overall response rate was identical in the two groups, at 71%, with continuous therapy generally well tolerated. Post-treatment pharmacokinetic assessments indicated full occupancy of BTK by ibrutinib at both dose levels, establishing 420 mg once daily as the standard dose in CLL/SLL. Importantly, the response rate of patients with deletions of the short arm of chromosome 17 (del17p) was comparable at 68%, highlighting the effectiveness of ibrutinib in patients who had historically had a poor prognosis with chemoimmunotherapy [7]. These initial findings were supported by the three RESONATE studies. The first RESONATE study demonstrated a significant improvement in both progression-free survival and overall survival (OS) with the use of ibrutinib when compared with the anti-CD20 antibody ofatumumab in patients with relapsed or refractory (R/R) CLL or SLL [8][9]. This was subsequently investigated in the frontline setting in the RESONATE-2 study, which also demonstrated a significant in improvement in PFS and OS with ibrutinib when compared with chlorambucil [10]. With a median follow-up of 60 months, PFS was 70% for ibrutinib versus 12% for chlorambucil, with ibrutinib also improving 5-year OS [11]. Ibrutinib was also well tolerated, with 42% of patients continuing to remain on ibrutinib at 8 years in a recently reported follow-up analysis [12]. Finally, the RESONATE-17 trial confirmed the efficacy of ibrutinib in patients with del17p, with 75% of patients remaining alive at 2 years—a significant improvement compared to historical controls [7][13]. While these initial trials could be criticized for comparing ibrutinib against relatively ineffective monotherapies, the superiority of ibrutinib has been confirmed by further studies versus chemoimmunotherapy. For example, the Eastern Cooperative Oncology Group–American College of Radiology Imaging Network (ECOG–ACRIN) study demonstrated an improvement in PFS and OS with ibrutinib–rituximab when compared with fludarabine, cyclophosphamide, and rituximab (FCR) [14][15]. Notably, a subgroup analysis suggested that the survival benefit is greater in patients with UM-IGHV genes, a finding replicated in other studies, logically suggesting that BTK inhibition is more effective in cases where the CLL cells have a higher BCR signalling capacity [16][17][18].
Initial studies also demonstrated the efficacy of the PI3K inhibitor, idelalisib, for the treatment of CLL [19]. A randomized phase 3 study compared idelalisib–rituximab to rituximab monotherapy in R/R CLL, showing an improvement in PFS and OS in the idelalisib arm [20]. While this led to the approval of idelalisib for CLL, it became apparent with further follow-up that this agent increases the incidence of several immune-related and infectious side effects including diarrhoea and colitis, hepatitis, CMV reactivation, and pneumonitis [21]. The higher incidence of adverse events was also observed with another PI3K inhibitor, duvelisib, suggesting a class effect and leading to black box warnings for both agents [22]. However, the efficacy of the idelalisib–rituximab combination does mean that it remains an option for some CLL patients, particularly those who are unsuitable for, or who are refractory to, BTKi- and venetoclax-based treatment [23][24]. Despite the fact that ibrutinib demonstrates a better safety profile than chemoimmunotherapy such as FCR, it is not without side effects. Common adverse events include diarrhoea, cough, infection, and myalgia, with bleeding another frequent and sometimes severe problem. One of the particular areas of concern with this agent is its cardiovascular effects: hypertension, atrial fibrillation, ventricular arrhythmia, and sudden cardiac death [25][26]. A critical feature is that while ibrutinib is an effective inhibitor of BTK, it also inhibits a wide variety of other kinases such as interleukin-2-inducible T-cell kinase and epidermal growth factor receptor tyrosine kinase [27]. Notably, it is ibrutinib’s inhibition of another kinase, C-terminal SRC kinase, that is thought to be responsible for the increased risk of atrial fibrillation seen with this drug [28]. As patients will need to take a BTKi continuously long-term to maintain control of their disease, there was a strong rationale for the development of highly specific BTKis for the treatment of CLL.

2. Increasing Specificity for BTK: Acalabrutinib and Zanubrutinib

The advent of the second-generation BTKis acalabrutinib and zanubrutinib now provides additional options in the management of front-line and relapsed CLL. Acalabrutinib (dosed at 100 mg twice daily) is a highly selective irreversible covalent BTKi licensed as monotherapy in R/R CLL following the ASCEND trial which demonstrated its superiority over investigator choice of either idelalisib–rituximab or bendamustine–rituximab [29][30]. Acalabrutinib is also now approved with/without the anti-CD20 monoclonal antibody obinutuzumab in the front-line setting following the ELEVATE-TN trial [31]. Both acalabrutinib monotherapy and acalabrutinib–obinutuzumab showed a substantial, superior 5-year PFS advantage compared with chlorambucil–obinutuzumab [32].
The ELEVATE-RR open-labelled randomized controlled trial directly compared acalabrutinib with ibrutinib in R/R CLL patients and at least one high-risk genetic aberrancy (del17p/TP53 mutation or 11q deletion) [33]. The study was designed to assess the noninferiority of PFS and also a hierarchical superiority assessment of toxicity differences between the two BTKis. At a median follow up of 40.9 months, there was no difference in PFS between the two agents but acalabrutinib-treated patients experienced a broad improvement in safety profile. All-grade cardiac adverse events and noncardiac adverse events (diarrhoea, myalgia/arthralgia, bleeding) were all improved with acalabrutinib with fewer adverse events leading to discontinuation. In light of this, the improved specificity of acalabrutinib for BTK does appear to translate into an improved safety profile when compared to ibrutinib, while retaining comparable efficacy. Notably, acalabrutinib has also demonstrated efficacy and safety in ibrutinib-intolerant CLL patients and is a valuable treatment option in this specific patient group [34][35].
Zanubrutinib (dosed at 160 mg twice daily) is another second-generation irreversible covalent BTKi approved as monotherapy for both front-line and relapsed CLL following results of the SEQUOIA trial and the ALPINE trial, respectively [36][37]. Zanubrutinib was developed to ensure better BTK specificity than ibrutinib and more sustained BTK occupancy, with exposure coverage above the half-maximal inhibitory concentration (IC50) across a 24 h dosing period [38]. The SEQUOIA trial demonstrated an improved PFS for continuous zanubrutinib compared to fixed-duration bendamustine–rituximab in patients in the front-line setting without del17p [37]. At a median follow up of 26.2 months, the median PFS was not reached in either group, but the 24-month PFS was 69.5% and 85.5%, respectively. The benefit was once again primarily seen in patients with UM-IGHV CLL. One hundred and ten patients with del17p/TP53 mutation were enrolled in a separate open label phase II nonrandomized arm of the SEQUOIA trial (Arm C), receiving zanubrutinib monotherapy to progression. At a median follow-up of 30.5 months, the overall response rate (ORR) was 90%, the estimated 24-month PFS was 88.9%, and the estimated 24-month overall survival was 93.6%.
The recently published ALPINE trial directly compared zanubrutinib monotherapy with ibrutinib monotherapy in a large open-label, phase 3, randomized trial with ORR (excluding partial response with lymphocytosis) the primary end point, with PFS and safety key secondary endpoints [36]. The population was broader than in ELEVATE-TN, with all BTKi-naïve relapsed CLL patients enrolled, and was a relatively low-risk patient cohort (median number of prior lines 1, TP53 mutated/17p deletion 23%). With a median follow-up of 29.6 months, zanubrutinib demonstrated a superior PFS compared to ibrutinib. This difference was also noted in a subgroup of patients with TP53 mutated/del17p CLL (HR 0.53). Discontinuation rates for reasons other than progression, cardiac sudden deaths, and atrial fibrillation rates were all important findings in favour of zanubrutinib. The toxicity profile between the two agents for other parameters such as hypertension, infection, bleeding, and cytopenia rates were similar. Zanubrutinib also recently demonstrated safety and efficacy in 67 patients previously intolerant to either ibrutinib or acalabrutinib [39].
As a result, it now seems reasonable to recommend these second-generation molecules with increased specificity for BTK over ibrutinib, due to the improved safety profile with acalabrutinib and the demonstrated improved efficacy and safety with zanubrutinib. Whether zanubrutinib is superior in terms of efficacy compared to acalabrutinib is unknown, and there are no head-to-head clinical trials planned or enrolling. The ALPINE and ELEVATE-RR trials studied different patient risk populations, with different geographies, across different treatment eras, and it is therefore impossible to cross-compare these studies. However, now that the clinical benefits of targeting BTK with improved specificity are proven, the next step in the evolutionary process of inhibiting this kinase in CLL is to test whether altering the mode of binding of drugs to BTK can further enhance the “fitness” of these agents for treating patients.

3. Noncovalent BTK Inhibition: Pirtobrutinib and Nemtabrutinib

While covalent BTKis have dramatically improved outcomes for patients with CLL/SLL, they are not curative. Long-term continuous usage can be associated with side effects that are difficult for patients to tolerate and also result in resistance due to development of mutations. Several resistance mutations have now been described, including a cysteine-to-serine mutation in BTK at the binding site of ibrutinib (C481S mutation) or gain-of-function mutations in the downstream kinase PLC-γ2 (R665W and L845F mutations) [40][41]. As a result, noncovalent BTKis have been developed with the potential to overcome the most common resistance mechanism associated with covalent BTKi use. Noncovalent binding to BTK does not rely on the C481 binding site, and so drugs can be designed that inhibit wildtype and C481-mutant BTK with equivalent potency. The two agents that are most advanced in clinical development are pirtobrutinib and nemtabrutinib.
Pirtobrutinib (formally LOXO-305) is a highly selective, first-in-class noncovalent (reversible) BTKi evaluated for both safety and efficacy in patients with CLL/SLL previously treated with a covalent BTKi in the first-in-human phase 1–2 BRUIN trial [42]. Pirtobrutinib has a favourable pharmacokinetic profile with high oral bioavailability and an extended half-life (approximately 19 h). This enables once-daily dosing with sustained plasma drug levels throughout the 24 h dosing interval, regardless of the intrinsic rate of BTK turnover. The selectivity profile of pirtobrutinib has the potential to minimize off-target inhibition and associated toxicity. The most recent update of the CLL patients in the BRUIN trial provided survival and toxicity data on 247 BTKi pretreated CLL/SLL patients with a median of 19.4 months follow-up [43]. The ORR for all patients was 82.2% when including partial response with lymphocytosis. Similar response rates were seen in dual-exposed (covalent BTKi and BCL2i) patients, TP53 aberrant patients, pentad-exposed patients (anti-CD20, chemotherapy, covalent BTKi, BCL2i, Pi3Ki), and C481-mutated patients. Perhaps unsurprisingly, response rates in patients with downstream mutations in PLCγ2 were lower (ORR 55.6%). The median PFS for the whole cohort was 19.6 months. The most common all-grade treatment-emergent adverse events (TEAEs) across all 773 B-cell malignancy patients in BRUIN treated with pirtobrutinib were fatigue (29%), neutropenia (24%), and diarrhoea (24%). The rates of BTKi-associated events of special interest were low, including hypertension (9.2%), atrial fibrillation/flutter (2.8%), minor bleeding, and major haemorrhage (2.2%) [44]. Notably, only 3% of patients across the whole of the BRUIN trial discontinued pirtobrutinib due to a treatment-related side effect. Pirtobrutinib also demonstrated an excellent safety profile in 123 patients with B-cell malignancies who stopped a prior covalent BTKi (ibrutinib (n = 118, 96%), acalabrutinib (n = 29, 24%), or zanubrutinib (n = 6, 5%)) due to intolerance. Overall, 7% of these 123 patients subsequently discontinued pirtobrutinib for adverse events (only four stopped for reasons related to pirtobrutinib), and recurrences of adverse events were generally at a lesser grade [45]. A number of ongoing randomized trials are enrolling to consolidate its role in covalent BTKi-exposed patients and potentially move the agent further up the treatment pathway, such as the BRUIN-322 study, which is comparing venetoclax–rituximab against the combination of venetoclax–rituximab–pirtobrutinib in BTK naïve and exposed patients [46].
Nemtabrutinib (MK-1026, formerly ARQ-531) is another noncovalent BTK in earlier development in the phase I-II 1/2 BELLWAVE-001 study. Recently updated efficacy data for 57 CLL/SLL pts with CLL/SLL treated with nemtabrutinib 65 mg once daily and safety for all 112 patients with B-cell malignancies treated at the 65 mg dose were presented [47]. The median number of prior lines of treatment was 4 (1–18), with 95% of those enrolled having received a prior covalent BTKi, with 42% also having received a BCL2 inhibitor. At a relatively short median follow-up of 8.1 months, the ORR was 56% and estimated median duration of response was 24.4 months. Seventy-three percent of patients experienced any-grade treatment-related adverse events. The most common (≥10%) were dysgeusia (21%), neutropenia (20%), fatigue (13%), nausea and thrombocytopenia (12% each), and diarrhoea and hypertension (10% each). Treatment-related discontinuations occurred in 15 pts (13%), somewhat higher than seen in pirtobrutinib-treated patients in the BRUIN trial (2.6%). Nemtabrutinib is less selective than pirtobrutinib, and further follow-up and a larger sample size is required to understand the future role of this promising agent.

4. BTK-Degradation: NX-2127 and NX-5948

Another BTK-targeting approach that may prove to be a critical addition to the CLL armamentarium is the strategy of degrading this kinase. In contrast to the agents discussed above which inhibit BTK function, “BTK degraders” essentially remove the BTK protein from the cell by targeting it for degradation by the proteasome. Ubiquitin-dependent proteolysis is a major pathway that degrades intracellular proteins as part of normal cellular maintenance processes [48]. In this pathway, proteins are targeted for degradation by the proteasome by the transfer of ubiquitin molecules to the protein of interest (in this case, BTK) by the combined action of ubiquitin-activating enzymes, ubiquitin-conjugating enzymes, and E3 ubiquitin–protein ligases. The first approaches utilized an existing drug (e.g., the angiogenesis inhibitor ovalicin) attached to short phosphopeptides that could be recognized by one of these E3 ubiquitin–protein ligases [49]. A major advance was the discovery that another E3 ubiquitin ligase, cereblon, was the target of thalidomide and its analogies lenalidomide and pomalidomide, widely used for the treatment of multiple myeloma [50][51]. These agents modulate cereblon to target IKAROS Family Zinc Finger 1 (IKZF1) and IKZF3 for degradation resulting in their immunomodulatory and anticancer activity. Several BTK degraders are being developed, including NX-2127 and NX-5948, which have now entered early phase clinical trials. Notably, NX-2127 targets both BTK and IKZF3 while NX-5948 just selectively degrades BTK. Both are currently in phase 1 studies with NX-2127 already demonstrating clinical responses in heavily pretreated (median 6 prior therapies) patients with CLL, including those with BTK mutations resistant to both covalent and noncovalent BTKis [52]. The ability of these drugs to potentially overcome the resistance mutations that emerge with BTK inhibitor treatment may mean that they form an important component of future treatment algorithms for CLL [53].

References

  1. Honigberg, L.A.; Smith, A.M.; Sirisawad, M.; Verner, E.; Loury, D.; Chang, B.; Li, S.; Pan, Z.; Thamm, D.H.; Miller, R.A.; et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc. Natl. Acad. Sci. USA 2010, 107, 13075–13080.
  2. Bruton, O.C. Agammaglobulinemia. Pediatrics 1952, 9, 722–728.
  3. Bruton, O.C.; Apt, L.; Gitlin, D.; Janeway, C.A. Absence of serum gamma globulins. AMA Am. J. Dis. Child. 1952, 84, 632–636.
  4. Rawlings, D.J.; Saffran, D.C.; Tsukada, S.; Largaespada, D.A.; Grimaldi, J.C.; Cohen, L.; Mohr, R.N.; Bazan, J.F.; Howard, M.; Copeland, N.G.; et al. Mutation of unique region of Bruton’s tyrosine kinase in immunodeficient XID mice. Science 1993, 261, 358–361.
  5. Herman, S.E.; Gordon, A.L.; Hertlein, E.; Ramanunni, A.; Zhang, X.; Jaglowski, S.; Flynn, J.; Jones, J.; Blum, K.A.; Buggy, J.J.; et al. Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood 2011, 117, 6287–6296.
  6. Byrd, J.C.; Furman, R.R.; Coutre, S.E.; Flinn, I.W.; Burger, J.A.; Blum, K.A.; Grant, B.; Sharman, J.P.; Coleman, M.; Wierda, W.G.; et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 2013, 369, 32–42.
  7. Hallek, M.; Fischer, K.; Fingerle-Rowson, G.; Fink, A.M.; Busch, R.; Mayer, J.; Hensel, M.; Hopfinger, G.; Hess, G.; von Grunhagen, U.; et al. Addition of rituximab to fludarabine and cyclophosphamide in patients with chronic lymphocytic leukaemia: A randomised, open-label, phase 3 trial. Lancet 2010, 376, 1164–1174.
  8. Byrd, J.C.; Brown, J.R.; O’Brien, S.; Barrientos, J.C.; Kay, N.E.; Reddy, N.M.; Coutre, S.; Tam, C.S.; Mulligan, S.P.; Jaeger, U.; et al. Ibrutinib versus Ofatumumab in Previously Treated Chronic Lymphoid Leukemia. N. Engl. J. Med. 2014, 371, 213–223.
  9. Byrd, J.C.; Hillmen, P.; O’Brien, S.; Barrientos, J.C.; Reddy, N.M.; Coutre, S.; Tam, C.S.; Mulligan, S.P.; Jaeger, U.; Barr, P.M.; et al. Long-term follow-up of the RESONATE phase 3 trial of ibrutinib vs ofatumumab. Blood 2019, 133, 2031–2042.
  10. Burger, J.A.; Tedeschi, A.; Barr, P.M.; Robak, T.; Owen, C.; Ghia, P.; Bairey, O.; Hillmen, P.; Bartlett, N.L.; Li, J.; et al. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2015, 373, 2425–2437.
  11. 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.
  12. Barr, P.M.; Owen, C.; Robak, T.; Tedeschi, A.; Bairey, O.; Burger, J.A.; Hillmen, P.; Coutre, S.E.; Dearden, C.; Grosicki, S.; et al. Up to 8-year follow-up from RESONATE-2: First-line ibrutinib treatment for patients with chronic lymphocytic leukemia. Blood Adv. 2022, 6, 3440–3450.
  13. O’Brien, S.; Jones, J.A.; Coutre, S.E.; Mato, A.R.; Hillmen, P.; Tam, C.; Osterborg, A.; Siddiqi, T.; Thirman, M.J.; Furman, R.R.; et al. Ibrutinib for patients with relapsed or refractory chronic lymphocytic leukaemia with 17p deletion (RESONATE-17): A phase 2, open-label, multicentre study. Lancet Oncol. 2016, 17, 1409–1418.
  14. Shanafelt, T.D.; Wang, X.V.; Kay, N.E.; Hanson, C.A.; O’Brien, S.; Barrientos, J.; Jelinek, D.F.; Braggio, E.; Leis, J.F.; Zhang, C.C.; et al. Ibrutinib-Rituximab or Chemoimmunotherapy for Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2019, 381, 432–443.
  15. Shanafelt, T.D.; Wang, X.V.; Hanson, C.A.; Paietta, E.M.; O’Brien, S.; Barrientos, J.; Jelinek, D.F.; Braggio, E.; Leis, J.F.; Zhang, C.C.; et al. Long-term outcomes for ibrutinib–rituximab and chemoimmunotherapy in CLL: Updated results of the E1912 trial. Blood 2022, 140, 112–120.
  16. Lanham, S.; Hamblin, T.; Oscier, D.; Ibbotson, R.; Stevenson, F.; Packham, G. Differential signaling via surface IgM is associated with VH gene mutational status and CD38 expression in chronic lymphocytic leukemia. Blood 2003, 101, 1087–1093.
  17. Mockridge, C.I.; Potter, K.N.; Wheatley, I.; Neville, L.A.; Packham, G.; Stevenson, F.K. Reversible anergy of sIgM-mediated signaling in the two subsets of CLL defined by VH-gene mutational status. Blood 2007, 109, 4424–4431.
  18. Moreno, C.; Greil, R.; Demirkan, F.; Tedeschi, A.; Anz, B.; Larratt, L.; Simkovic, M.; Samoilova, O.; Novak, J.; Ben-Yehuda, D.; et al. Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 43–56.
  19. Herman, S.E.; Gordon, A.L.; Wagner, A.J.; Heerema, N.A.; Zhao, W.; Flynn, J.M.; Jones, J.; Andritsos, L.; Puri, K.D.; Lannutti, B.J.; et al. Phosphatidylinositol 3-kinase-delta inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals. Blood 2010, 116, 2078–2088.
  20. Furman, R.R.; Sharman, J.P.; Coutre, S.E.; Cheson, B.D.; Pagel, J.M.; Hillmen, P.; Barrientos, J.C.; Zelenetz, A.D.; Kipps, T.J.; Flinn, I.; et al. Idelalisib and Rituximab in Relapsed Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2014, 370, 997–1007.
  21. Sharman, J.P.; Coutre, S.E.; Furman, R.R.; Cheson, B.D.; Pagel, J.M.; Hillmen, P.; Barrientos, J.C.; Zelenetz, A.D.; Kipps, T.J.; Flinn, I.W.; et al. Final Results of a Randomized, Phase III Study of Rituximab With or Without Idelalisib Followed by Open-Label Idelalisib in Patients With Relapsed Chronic Lymphocytic Leukemia. J. Clin. Oncol. 2019, 37, 1391–1402.
  22. Skanland, S.S.; Brown, J.R. PI3K inhibitors in chronic lymphocytic leukemia: Where do we go from here? Haematologica 2023, 108, 9–21.
  23. Walewska, R.; Parry-Jones, N.; Eyre, T.A.; Follows, G.; Martinez-Calle, N.; McCarthy, H.; Parry, H.; Patten, P.E.M.; Riches, J.C.; Hillmen, P.; et al. Guideline for the treatment of chronic lymphocytic leukaemia. Br. J. Haematol. 2022, 197, 544–557.
  24. Mato, A.R.; Roeker, L.E.; Jacobs, R.; Hill, B.T.; Lamanna, N.; Brander, D.; Shadman, M.; Ujjani, C.S.; Yazdy, M.S.; Perini, G.F.; et al. Assessment of the Efficacy of Therapies Following Venetoclax Discontinuation in CLL Reveals BTK Inhibition as an Effective Strategy. Clin. Cancer Res. 2020, 26, 3589–3596.
  25. Tang, C.P.S.; Lip, G.Y.H.; McCormack, T.; Lyon, A.R.; Hillmen, P.; Iyengar, S.; Martinez-Calle, N.; Parry-Jones, N.; Patten, P.E.M.; Schuh, A.; et al. Management of cardiovascular complications of bruton tyrosine kinase inhibitors. Br. J. Haematol. 2022, 196, 70–78.
  26. Munir, T.; Pitchford, A.; Bloor, A.; Broom, A.; Young, M.; Kennedy, B.; Walewska, R.; Furtado, M.; Preston, G.; Neilson, J.R.; et al. Sudden or Cardiac Deaths on Ibrutinib-Based Therapy Were Associated with a Prior History of Hypertension or Cardiac Disease and the Use of ACE-Inhibitors at Study Entry: Analysis from the Phase III NCRI FLAIR Trial. Blood 2021, 138 (Suppl. S1), 2636.
  27. Robak, T.; Witkowska, M.; Smolewski, P. The Role of Bruton’s Kinase Inhibitors in Chronic Lymphocytic Leukemia: Current Status and Future Directions. Cancers 2022, 14, 771.
  28. Xiao, L.; Salem, J.E.; Clauss, S.; Hanley, A.; Bapat, A.; Hulsmans, M.; Iwamoto, Y.; Wojtkiewicz, G.; Cetinbas, M.; Schloss, M.J.; et al. Ibrutinib-Mediated Atrial Fibrillation Attributable to Inhibition of C-Terminal Src Kinase. Circulation 2020, 142, 2443–2455.
  29. Ghia, P.; Pluta, A.; Wach, M.; Lysak, D.; Kozak, T.; Simkovic, M.; Kaplan, P.; Kraychok, I.; Illes, A.; de la Serna, J.; et al. ASCEND: Phase III, Randomized Trial of Acalabrutinib Versus Idelalisib Plus Rituximab or Bendamustine Plus Rituximab in Relapsed or Refractory Chronic Lymphocytic Leukemia. J. Clin. Oncol. 2020, 38, 2849–2861.
  30. Ghia, P.; Pluta, A.; Wach, M.; Lysak, D.; Simkovic, M.; Kriachok, I.; Illes, A.; de la Serna, J.; Dolan, S.; Campbell, P.; et al. Acalabrutinib Versus Investigator’s Choice in Relapsed/Refractory Chronic Lymphocytic Leukemia: Final ASCEND Trial Results. Hemasphere 2022, 6, e801.
  31. Sharman, J.P.; Egyed, M.; Jurczak, W.; Skarbnik, A.; Pagel, J.M.; Flinn, I.W.; Kamdar, M.; Munir, T.; Walewska, R.; Corbett, G.; 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, 1278–1291.
  32. Sharman, J.P.; Egyed, M.; Jurczak, W.; Skarbnik, A.P.; Kamdar, M.K.; Munir, T.; Fogliatto, L.; Herishanu, Y.; Banerji, V.; Follows, G.; et al. Acalabrutinib ± obinutuzumab versus obinutuzumab + chlorambucil in treatment-naïve chronic lymphocytic leukemia: Five-year follow-up of ELEVATE-TN. J. Clin. Oncol. 2022, 40, 7539.
  33. Byrd, J.C.; Hillmen, P.; Ghia, P.; Kater, A.P.; Chanan-Khan, A.; Furman, R.R.; O’Brien, S.; Yenerel, M.N.; Illés, A.; Kay, N.; et al. Acalabrutinib Versus Ibrutinib in Previously Treated Chronic Lymphocytic Leukemia: Results of the First Randomized Phase III Trial. J. Clin. Oncol. 2021, 39, 3441–3452.
  34. Rogers, K.A.; Thompson, P.A.; Allan, J.N.; Coleman, M.; Sharman, J.P.; Cheson, B.D.; Jones, D.; Izumi, R.; Frigault, M.M.; Quah, C.; et al. Phase II study of acalabrutinib in ibrutinib-intolerant patients with relapsed/refractory chronic lymphocytic leukemia. Haematologica 2021, 106, 2364–2373.
  35. Awan, F.T.; Schuh, A.; Brown, J.R.; Furman, R.R.; Pagel, J.M.; Hillmen, P.; Stephens, D.M.; Woyach, J.; Bibikova, E.; Charuworn, P.; et al. Acalabrutinib monotherapy in patients with chronic lymphocytic leukemia who are intolerant to ibrutinib. Blood Adv. 2019, 3, 1553–1562.
  36. Brown, J.R.; Eichhorst, B.; Hillmen, P.; Jurczak, W.; Kaźmierczak, M.; Lamanna, N.; O’Brien, S.M.; Tam, C.S.; Qiu, L.; Zhou, K.; et al. Zanubrutinib or Ibrutinib in Relapsed or Refractory Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2022, 388, 319–332.
  37. Tam, C.S.; Brown, J.R.; Kahl, B.S.; Ghia, P.; Giannopoulos, K.; Jurczak, W.; Simkovic, M.; Shadman, M.; Osterborg, A.; Laurenti, L.; et al. Zanubrutinib versus bendamustine and rituximab in untreated chronic lymphocytic leukaemia and small lymphocytic lymphoma (SEQUOIA): A randomised, controlled, phase 3 trial. Lancet Oncol. 2022, 23, 1031–1043.
  38. Tam, C.S.; Trotman, J.; Opat, S.; Burger, J.A.; Cull, G.; Gottlieb, D.; Harrup, R.; Johnston, P.B.; Marlton, P.; Munoz, J.; et al. Phase 1 study of the selective BTK inhibitor zanubrutinib in B-cell malignancies and safety and efficacy evaluation in CLL. Blood 2019, 134, 851–859.
  39. Shadman, M.; Flinn, I.W.; Levy, M.Y.; Porter, R.F.; Burke, J.M.; Zafar, S.F.; Misleh, J.; Kingsley, E.C.; Yimer, H.A.; Freeman, B.; et al. Zanubrutinib in patients with previously treated B-cell malignancies intolerant of previous Bruton tyrosine kinase inhibitors in the USA: A phase 2, open-label, single-arm study. Lancet Haematol. 2023, 10, e35–e45.
  40. Woyach, J.; Huang, Y.; Rogers, K.; Bhat, S.A.; Grever, M.R.; Lozanski, A.; Doong, T.-J.; Blachly, J.S.; Lozanski, G.; Jones, D.; et al. Resistance to Acalabrutinib in CLL Is Mediated Primarily By BTK Mutations. Blood 2019, 134, 504.
  41. Woyach, J.A.; Furman, R.R.; Liu, T.-M.; Ozer, H.G.; Zapatka, M.; Ruppert, A.S.; Xue, L.; Li, D.H.-H.; Steggerda, S.M.; Versele, M.; et al. Resistance Mechanisms for the Bruton’s Tyrosine Kinase Inhibitor Ibrutinib. N. Engl. J. Med. 2014, 370, 2286–2294.
  42. Mato, A.R.; Shah, N.N.; Jurczak, W.; Cheah, C.Y.; Pagel, J.M.; Woyach, J.A.; Fakhri, B.; Eyre, T.A.; Lamanna, N.; Patel, M.R.; et al. Pirtobrutinib in relapsed or refractory B-cell malignancies (BRUIN): A phase 1/2 study. Lancet 2021, 397, 892–901.
  43. Mato, A.R.; Woyach, J.A.; Brown, J.R.; Ghia, P.; Patel, K.; Eyre, T.A.; Munir, T.; Lech-Marańda, E.; Lamanna, N.; Tam, C.S.; et al. Efficacy of Pirtobrutinib in Covalent BTK-Inhibitor Pre-Treated Relapsed / Refractory CLL/SLL: Additional Patients and Extended Follow-up from the Phase 1/2 BRUIN Study. Blood 2022, 140 (Suppl. S1), 2316–2320.
  44. Bye, A.P.; Kriek, N.; Sage, T.; Rawlings, S.J.; Prodger, C.; Kesavan, M.; Lees, C.; Booth, S.; Cowen, L.G.; Shefferd, K.; et al. Pirtobrutinib results in reversible platelet dysfunction compared to ibrutinib and acalabrutinib. Haematologica 2022, 108, 1429–1435.
  45. Shah, N.N.; Wang, M.L.; Brown, J.R.; Patel, K.; Woyach, J.A.; Wierda, W.G.; Ujjani, C.S.; Eyre, T.A.; Zinzani, P.L.; Alencar, A.J.; et al. Safety and Tolerability of Pirtobrutinib Monotherapy in Patients with B-Cell Malignancies Who Were Previously Intolerant to a Covalent BTK Inhibitor: Results from the Phase 1/2 BRUIN Study. Blood 2022, 140 (Suppl. S1), 4127–4132.
  46. Mato, A.R.; Wierda, W.G.; Pagel, J.M.; Davids, M.S.; Zinzani, P.L.; Lu, Y.; Liu, H.; Shahda, S.; Leow, C.C.; Tam, C.S.; et al. BRUIN CLL-322: A Phase 3 Open-Label, Randomized Study of Fixed Duration Pirtobrutinib Plus Venetoclax and Rituximab Versus Venetoclax and Rituximab in Previously Treated Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma (Trial in Progress). Blood 2021, 138, 3742.
  47. Woyach, J.A.; Flinn, I.W.; Awan, F.T.; Eradat, H.; Brander, D.; Tees, M.; Parikh, S.A.; Phillips, T.J.; Ghori, R.; Reddy, N.M.; et al. Efficacy and Safety of Nemtabrutinib, a Wild-Type and C481S-Mutated Bruton Tyrosine Kinase Inhibitor for B-Cell Malignancies: Updated Analysis of the Open-Label Phase 1/2 Dose-Expansion Bellwave-001 Study. Blood 2022, 140 (Suppl. S1), 7004–7006.
  48. Békés, M.; Langley, D.R.; Crews, C.M. PROTAC targeted protein degraders: The past is prologue. Nat. Rev. Drug Discov. 2022, 21, 181–200.
  49. Sakamoto, K.M.; Kim, K.B.; Kumagai, A.; Mercurio, F.; Crews, C.M.; Deshaies, R.J. Protacs: Chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation. Proc. Natl. Acad. Sci. USA 2001, 98, 8554–8559.
  50. Riches, J.C.; Gribben, J.G. Mechanistic and Clinical Aspects of Lenalidomide Treatment for Chronic Lymphocytic Leukemia. Curr. Cancer Drug Targets 2016, 16, 689–700.
  51. Ito, T.; Ando, H.; Suzuki, T.; Ogura, T.; Hotta, K.; Imamura, Y.; Yamaguchi, Y.; Handa, H. Identification of a Primary Target of Thalidomide Teratogenicity. Science 2010, 327, 1345–1350.
  52. Mato, A.R.; Wierda, W.G.; Ai, W.Z.; Flinn, I.W.; Tees, M.; Patel, M.R.; Patel, K.; O’Brien, S.; Bond, D.A.; Roeker, L.E.; et al. NX-2127-001, a First-in-Human Trial of NX-2127, a Bruton’s Tyrosine Kinase-Targeted Protein Degrader, in Patients with Relapsed or Refractory Chronic Lymphocytic Leukemia and B-Cell Malignancies. Blood 2022, 140 (Suppl. S1), 2329–2332.
  53. Montoya, S.; Bourcier, J.; Thompson, M.C.; Noviski, M.; Tan, M.; Wang, E.; Mi, X.; Brathaban, N.; Barrientos Risso, C.; Tsai, D.; et al. Kinase Dead BTK Mutations Confer Resistance to Covalent and Noncovalent BTK Inhibitors but Are Susceptible to Clinical Stage BTK Degraders. Blood 2022, 140 (Suppl. S1), 1811–1813.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 199
Revisions: 2 times (View History)
Update Date: 19 May 2023
1000/1000
Video Production Service