Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

Molecular targets for therapy

Pharmacodynamics and proteomic analysis of acalabrutinib therapy: similarity of on-target effects to ibrutinib and rationale for combination therapy

Abstract

Acalabrutinib, a highly selective Bruton’s tyrosine kinase inhibitor, is associated with high overall response rates and durable remission in previously treated chronic lymphocytic leukemia (CLL); however, complete remissions were limited. To elucidate on-target and pharmacodynamic effects of acalabrutinib, we evaluated several laboratory endpoints, including proteomic changes, chemokine modulation and impact on cell migration. Pharmacological profiling of samples from acalabrutinib-treated CLL patients was used to identify strategies for achieving deeper responses, and to identify additive/synergistic combination regimens. Peripheral blood samples from 21 patients with relapsed/refractory CLL in acalabrutinib phase I (100–400 mg/day) and II (100 mg BID) clinical trials were collected prior to and on days 8 and 28 after treatment initiation and evaluated for plasma chemokines, reverse phase protein array, immunoblotting and pseudoemperipolesis. The on-target pharmacodynamic profile of acalabrutinib in CLL lymphocytes was comparable to ibrutinib in measures of acalabrutinib-mediated changes in CCL3/CCL4 chemokine production, migration assays and changes in B-cell receptor signaling pathway proteins and other downstream survival proteins. Among several CLL-targeted agents, venetoclax, when combined with acalabrutinib, showed optimal complementary activity in vitro, ex vivo and in vivo in TCL-1 adoptive transfer mouse model system of CLL. These findings support selective targeting and combinatorial potential of acalabrutinib.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7
Figure 8

Similar content being viewed by others

References

  1. Hendriks RW, Yuvaraj S, Kil LP . Targeting Bruton's tyrosine kinase in B cell malignancies. Nat Rev Cancer 2014; 14: 219–232.

    Article  CAS  PubMed  Google Scholar 

  2. Stevenson FK, Caligaris-Cappio F . Chronic lymphocytic leukemia: revelations from the B-cell receptor. Blood 2004; 103: 4389–4395.

    Article  CAS  PubMed  Google Scholar 

  3. Ponader S, Burger JA . Bruton's tyrosine kinase: from X-linked agammaglobulinemia toward targeted therapy for B-cell malignancies. J Clin Oncol 2014; 32: 1830–1839.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wiestner A . Emerging role of kinase-targeted strategies in chronic lymphocytic leukemia. Blood 2012; 120: 4684–4691.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Byrd JC, Harrington B, O'Brien S, Jones JA, Schuh A, Devereux S et al. Acalabrutinib (ACP-196) in relapsed chronic lymphocytic leukemia. N Engl J Med 2016; 374: 323–332.

    Article  CAS  PubMed  Google Scholar 

  6. Honigberg LA, Smith AM, Sirisawad M, Verner E, Loury D, Chang B 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Herman SE, Gordon AL, Hertlein E, Ramanunni A, Zhang X, Jaglowski S 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Patel V, Balakrishnan K, Bibikova E, Ayres M, Keating MJ, Wierda WG et al. Comparison of acalabrutinib, a selective Bruton tyrosine kinase inhibitor, with ibrutinib in chronic lymphocytic leukemia cells. Clin Cancer Res 2017; 23: 3734–3743.

    Article  CAS  PubMed  Google Scholar 

  9. Dubovsky JA, Beckwith KA, Natarajan G, Woyach JA, Jaglowski S, Zhong Y et al. Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes. Blood 2013; 122: 2539–2549.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gao W, Wang M, Wang L, Lu H, Wu S, Dai B et al. Selective antitumor activity of ibrutinib in EGFR-mutant non-small cell lung cancer cells. J Natl Cancer Inst 2014; 106: dju204.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Byrd JC, Brown JR, O'Brien S, Barrientos JC, Kay NE, Reddy NM et al. Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med 2014; 371: 213–223.

    Article  PubMed  PubMed Central  Google Scholar 

  12. O'Brien S, Furman RR, Coutre SE, Sharman JP, Burger JA, Blum KA et al. Ibrutinib as initial therapy for elderly patients with chronic lymphocytic leukaemia or small lymphocytic lymphoma: an open-label, multicentre, phase 1b/2 trial. Lancet Oncol 2014; 15: 48–58.

    Article  CAS  PubMed  Google Scholar 

  13. O'Brien S, Jones JA, Coutre SE, Mato AR, Hillmen P, Tam C 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.

    Article  CAS  PubMed  Google Scholar 

  14. Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med 2015; 373: 2425–2437.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Burger JA, Burger M, Kipps TJ . Chronic lymphocytic leukemia B cells express functional CXCR4 chemokine receptors that mediate spontaneous migration beneath bone marrow stromal cells. Blood 1999; 94: 3658–3667.

    CAS  PubMed  Google Scholar 

  16. Balakrishnan K, Peluso M, Fu M, Rosin NY, Burger JA, Wierda WG et al. The phosphoinositide-3-kinase (PI3K)-delta and gamma inhibitor, IPI-145 (Duvelisib), overcomes signals from the PI3K/AKT/S6 pathway and promotes apoptosis in CLL. Leukemia 2015; 29: 1811–1822.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Burger JA, Quiroga MP, Hartmann E, Burkle A, Wierda WG, Keating MJ et al. High-level expression of the T-cell chemokines CCL3 and CCL4 by chronic lymphocytic leukemia B cells in nurselike cell cocultures and after BCR stimulation. Blood 2009; 113: 3050–3058.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen SS, Batliwalla F, Holodick NE, Yan XJ, Yancopoulos S, Croce CM et al. Autoantigen can promote progression to a more aggressive TCL1 leukemia by selecting variants with enhanced B-cell receptor signaling. Proc Natl Acad Sci USA 2013; 110: E1500–E1507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Herman SE, Mustafa RZ, Jones J, Wong DH, Farooqui M, Wiestner A . Treatment with ibrutinib inhibits BTK- and VLA-4-dependent adhesion of chronic lymphocytic leukemia cells in vivo. Clin Cancer Res 2015; 21: 4642–4651.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ponader S, Chen SS, Buggy JJ, Balakrishnan K, Gandhi V, Wierda WG et al. The Bruton tyrosine kinase inhibitor PCI-32765 thwarts chronic lymphocytic leukemia cell survival and tissue homing in vitro and in vivo. Blood 2012; 119: 1182–1189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Niemann CU, Herman SE, Maric I, Gomez-Rodriguez J, Biancotto A, Chang BY et al. 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: 1572–1582.

    Article  CAS  PubMed  Google Scholar 

  22. Kohrt HE, Sagiv-Barfi I, Rafiq S, Herman SE, Butchar JP, Cheney C et al. Ibrutinib antagonizes rituximab-dependent NK cell-mediated cytotoxicity. Blood 2014; 123: 1957–1960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fiorcari S, Maffei R, Audrito V, Martinelli S, Ten Hacken E, Zucchini P et al. Ibrutinib modifies the function of monocyte/macrophage population in chronic lymphocytic leukemia. Oncotarget 2016; 7: 65968–65981.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Ren L, Campbell A, Fang H, Gautam S, Elavazhagan S, Fatehchand K et al. Analysis of the effects of the Bruton's tyrosine kinase (Btk) inhibitor ibrutinib on monocyte Fcgamma receptor (FcgammaR) function. J Biol Chem 2016; 291: 3043–3052.

    Article  CAS  PubMed  Google Scholar 

  25. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA 2002; 99: 15524–15529.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA 2005; 102: 13944–13949.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Kitada S, Andersen J, Akar S, Zapata JM, Takayama S, Krajewski S et al. Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with In vitro and In vivo chemoresponses. Blood 1998; 91: 3379–3389.

    CAS  PubMed  Google Scholar 

  28. Balakrishnan K, Burger JA, Fu M, Doifode T, Wierda WG, Gandhi V . Regulation of Mcl-1 expression in context to bone marrow stromal microenvironment in chronic lymphocytic leukemia. Neoplasia 2014; 16: 1036–1046.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Herndon TM, Chen SS, Saba NS, Valdez J, Emson C, Gatmaitan M et al. Direct in vivo evidence for increased proliferation of CLL cells in lymph nodes compared to bone marrow and peripheral blood. Leukemia 2017; 31: 1340–1347.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 2005; 435: 677–681.

    Article  CAS  PubMed  Google Scholar 

  31. Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med 2013; 19: 202–208.

    Article  CAS  PubMed  Google Scholar 

  32. Niemann CU, Mora-Jensen HI, Dadashian EL, Krantz F, Covey T, Chen SS et al. Combined BTK and PI3Kdelta inhibition with acalabrutinib and ACP-319 improves survival and tumor control in CLL mouse model. Clin Cancer Res 2017; 23: 5814–5823.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cervantes-Gomez F, Lamothe B, Woyach JA, Wierda WG, Keating MJ, Balakrishnan K et al. Pharmacological and protein profiling suggests venetoclax (ABT-199) as optimal partner with ibrutinib in chronic lymphocytic leukemia. Clin Cancer Res 2015; 21: 3705–3715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yang Q, Shah P, Korkut A, Fernandes SM, Hanna J, Brown JR et al. Changes in Bcl-2 family protein profile during idelalisib therapy mimic those during duvelisib therapy in CLL lymphocytes. JCO Precis Oncol 2017; 1: 1–5.

    CAS  Google Scholar 

  35. Patel VM, Balakrishnan K, Douglas M, Tibbitts T, Xu EY, Kutok JL et al. Duvelisib treatment is associated with altered expression of apoptotic regulators that helps in sensitization of chronic lymphocytic leukemia cells to venetoclax (ABT-199). Leukemia 2017; 31: 1872–1881.

    Article  CAS  PubMed  Google Scholar 

  36. Deng J, Isik E, Fernandes SM, Brown JR, Letai A, Davids MS . Bruton's tyrosine kinase inhibition increases BCL-2 dependence and enhances sensitivity to venetoclax in chronic lymphocytic leukemia. Leukemia 2017; 31: 2075–2208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Maurer U, Charvet C, Wagman AS, Dejardin E, Green DR . Glycogen synthase kinase-3 regulates mitochondrial outer membrane permeabilization and apoptosis by destabilization of MCL-1. Mol Cell 2006; 21: 749–760.

    Article  CAS  PubMed  Google Scholar 

  38. Xu C, Kim NG, Gumbiner BM . Regulation of protein stability by GSK3 mediated phosphorylation. Cell Cycle 2009; 8: 4032–4039.

    Article  CAS  PubMed  Google Scholar 

  39. Kang T, Wei Y, Honaker Y, Yamaguchi H, Appella E, Hung MC et al. GSK-3 beta targets Cdc25A for ubiquitin-mediated proteolysis, and GSK-3 beta inactivation correlates with Cdc25A overproduction in human cancers. Cancer Cell 2008; 13: 36–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bhanot H, Reddy MM, Nonami A, Weisberg EL, Bonal D, Kirschmeier PT et al. Pathological glycogenesis through glycogen synthase 1 and suppression of excessive AMP kinase activity in myeloid leukemia cells. Leukemia 2015; 29: 1555–1563.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Greer EL, Brunet A . FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 2005; 24: 7410–7425.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are thankful to Ingrid Chou Koo, PhD, Team 9, for critically editing the manuscript and for her help with references; and to Ningping Feng, Xiaoyan Ma, Andy Zuniga and the in vivo pharmacology team at the Center for Co-Clinical Trials at MD Anderson Cancer Center for their expert assistance for in vivo experiments. We are also grateful to Todd Covey and Allard Kaptein, Acerta Pharma for critically reviewing the manuscript and providing constructive scientific comments. This work was supported by a Sponsored Research Agreement from Acerta and philanthropic funds from the MD Anderson’s CLL Moon Shot program. MD Anderson Cancer Center is supported in part by the National Institutes of Health through Cancer Center Support Grant P30CA016672.

Author contributions

VKP designed and performed the experiments, analyzed the results, and wrote portion of the manuscript. BL established adoptive transfer mouse model and performed mice in vivo experiments and assays, wrote portion of the manuscript. MLA assisted in experimental planning and performing immunoblots. JG performed in vivo experiments. JPC did BTK occupancy assays at Acerta Pharma. KB directed CCL3/4 assays and wrote parts of the manuscript. CI analyzed RPPA data. JM as a summer student performed in vitro combination experiments. MN identified patient samples for in vitro studies and provided patient characteristics. MJK and WGW identified patients to obtain peripheral blood samples, provided clinical and patient-related input, and reviewed the manuscript. JRM supervised mouse model investigations. VG conceptualized and supervised the research, obtained funding, analyzed the data, and wrote majority of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to V Gandhi.

Ethics declarations

Competing interests

VG and WGW received research and clinical trial funding from Acerta. JPC is an employee of Acerta. The other authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on the Leukemia website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Patel, V., Lamothe, B., Ayres, M. et al. Pharmacodynamics and proteomic analysis of acalabrutinib therapy: similarity of on-target effects to ibrutinib and rationale for combination therapy. Leukemia 32, 920–930 (2018). https://doi.org/10.1038/leu.2017.321

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/leu.2017.321

This article is cited by

Search

Quick links