PI3Kδ inhibitor idelalisib in combination with BTK inhibitor ONO/GS-4059 in diffuse large B cell lymphoma with acquired resistance to PI3Kδ and BTK inhibitors

Anella Yahiaoui, Sarah A Meadows, Rick A Sorensen, Zhi-Hua Cui, Kathleen S Keegan, Robert Brockett, Guang Chen, Christophe Quéva, Li Li, Stacey L Tannheimer, Anella Yahiaoui, Sarah A Meadows, Rick A Sorensen, Zhi-Hua Cui, Kathleen S Keegan, Robert Brockett, Guang Chen, Christophe Quéva, Li Li, Stacey L Tannheimer

Abstract

Activated B-cell-like diffuse large B-cell lymphoma relies on B-cell receptor signaling to drive proliferation and survival. Downstream of the B-cell receptor, the key signaling kinases Bruton's tyrosine kinase and phosphoinositide 3-kinase δ offer opportunities for therapeutic intervention by agents such as ibrutinib, ONO/GS-4059, and idelalisib. Combination therapy with such targeted agents could provide enhanced efficacy due to complimentary mechanisms of action. In this study, we describe both the additive interaction of and resistance mechanisms to idelalisib and ONO/GS-4059 in a model of activated B-cell-like diffuse large B-cell lymphoma. Significant tumor regression was observed with a combination of PI3Kδ and Bruton's tyrosine kinase inhibitors in the mouse TMD8 xenograft. Acquired resistance to idelalisib in the TMD8 cell line occurred by loss of phosphatase and tensin homolog and phosphoinositide 3-kinase pathway upregulation, but not by mutation of PIK3CD. Sensitivity to idelalisib could be restored by combining idelalisib and ONO/GS-4059. Further evaluation of targeted inhibitors revealed that the combination of idelalisib and the phosphoinositide-dependent kinase-1 inhibitor GSK2334470 or the AKT inhibitor MK-2206 could partially overcome resistance. Characterization of acquired Bruton's tyrosine kinase inhibitor resistance revealed a novel tumor necrosis factor alpha induced protein 3 mutation (TNFAIP3 Q143*), which led to a loss of A20 protein, and increased p-IκBα. The combination of idelalisib and ONO/GS-4059 partially restored sensitivity in this resistant line. Additionally, a mutation in Bruton's tyrosine kinase at C481F was identified as a mechanism of resistance. The combination activity observed with idelalisib and ONO/GS-4059, taken together with the ability to overcome resistance, could lead to a new therapeutic option in activated B-cell-like diffuse large B-cell lymphoma. A clinical trial is currently underway to evaluate the combination of idelalisib and ONO/GS-4059 (NCT02457598).

Conflict of interest statement

All authors were employees of the study sponsor, Gilead Sciences, Inc, at the time the study was conducted. Additionally, we adhere to the PLOS ONE guidelines on sharing data and materials.

Figures

Fig 1. TMD8 as a model for…
Fig 1. TMD8 as a model for evaluation of PI3Kδ and BTK inhibition.
(A) Cell viability with isoform-specific PI3K inhibitors was assessed by CellTiter Glo after 96 hours, mean ± SEM, n = 4. (B) PI3K isoform expression in TMD8 cells using Simple Western, concentration (pg/μL) quantitated by recombinant protein standards, representative experiment shown of n = 3. (C) Cell viability of TMD8 was assessed with idelalisib in combination with ONO/GS-4059 at fixed concentrations (3, 6, or 12 nM) or with ONO/GS-4059 in combination with idelalisib at fixed concentrations (20, 100 or 600 nM) after 96 hours, mean ± SEM, n = 8. (D) Cells were treated with idelalisib (420 nM), ONO/GS-4059 (320 nM) or in combination for 48 hours and apoptosis was assessed by FITC Annexin V staining, and measured by flow cytometry, mean values ± SD, n = 4. (E) Cells were treated with idelalisib (420 nM), ONO/GS-4059 (320 nM) or in combination for 2 and 24 hours; lysates were analyzed by western blot (p-AKT S473) and Simple Western, representative experiment shown of n = 3.
Fig 2. In vivo inhibition of PI3Kδ…
Fig 2. In vivo inhibition of PI3Kδ and BTK leads to TMD8 tumor regression.
(A) Response of randomized TMD8 xenograft tumors (n = 13 per group) treated with PI3Kδ inhibitor (GS-649443, 1 or 5 mg/kg, BID), BTK inhibitor (ONO/GS-4059, 3 or 10 mg/kg, BID) or combination. Tumor volumes are expressed as mean ± SEM with P

Fig 3. Combination of idelalisib and ONO/GS-4059…

Fig 3. Combination of idelalisib and ONO/GS-4059 can overcome TMD8 acquired idelalisib resistance.

(A) Cell…

Fig 3. Combination of idelalisib and ONO/GS-4059 can overcome TMD8 acquired idelalisib resistance.
(A) Cell viability of vehicle control line (TMD8IDELA-S) or idelalisib resistant line (TMD8IDELA-R) in response to idelalisib treatment, 96 hour CellTiterGlo assay, mean ± SEM, n = 4. (B) Cell viability of TMD8IDELA-S (top graph) or TMD8IDELA-R cells (bottom graph) treated with idelalisib, ONO/GS-4059 or ONO/GS-4059 in combination with idelalisib (1 μM), n = 4, mean ± SEM. Combination curve is normalized to the single agent alone. (C) Protein lysates from TMD8IDELA-S (vehicle treated) and TMD8IDELA-R treated with vehicle, idelalisib (420 nM), ONO/GS-4059 (320 nM) or in combination for 2 hours were generated and analyzed in a single run using Simple Western, representative image of a single run of n = 3. (D) Quantification of Fig 3C showing AUC normalization to actin, and values normalized to TMD8IDELA-S DMSO treated control for all treatment groups from a single run.

Fig 4. PI3K upregulation in acquired resistance…

Fig 4. PI3K upregulation in acquired resistance is sensitive to idelalisib and AKT or PDK1…

Fig 4. PI3K upregulation in acquired resistance is sensitive to idelalisib and AKT or PDK1 inhibitor combinations.
(A) Protein lysates were generated from TMD8IDELA-S and TMD8IDELA-R cells, and analyzed by western blot (p-AKT S473, p-AKT T308, AKT, p-S6RP S235/236, S6RP, p-GSK3β S9, GSK3β) and Simple Western (PTEN). (B) Cell viability of TMD8IDELA-S (left) and TMD8IDELA-R (right) cells treated with idelalisib, MK-2206 or combination of idelalisib and MK-2206 (1 μM), 96 hours CellTiterGlo assay, mean ± SEM, n = 4. Combination curve is normalized to the single agent alone. (C) Cell viability of TMD8IDELA-S (left) and TMD8IDELA-R (right) cells treated with idelalisib, GSK2334470 or combination of idelalisib and GSK2334470 (3 μM), 96 hours CellTiterGlo assay, mean ± SEM, n = 4. Combination curve is normalized to the single agent alone. (D) TMD8IDELA-R cells were treated with vehicle, idelalisib (1 μM), MK-2206 (1 μM), and idelalisib plus MK-2206, or (E) Vehicle, idelalisib (1 μM), GSK2334470 (1 μM), and idelalisib plus GSK2334470 for 48 hours. Apoptosis was assessed by 7AAD and PE annexin V staining, and analyzed by flow cytometry, mean ± SD, n = 3. (F) TMD8IDELA-R cells were treated with vehicle, idelalisib (1 μM), MK-2206 (1 μM), or idelalisib plus MK-2206 and (G) Vehicle, idelalisib (1 μM), GSK2334470 (1 μM), or idelalisib plus GSK2334470 for 2 hours. Protein lysates were generated and analyzed by western blot, representative experiment of n = 3. TMD8IDELA-S vehicle control was included as a reference on the same membrane.

Fig 5. Loss of A20 and BTK…

Fig 5. Loss of A20 and BTK C481F mutation as mechanisms of BTK inhibitor resistance…

Fig 5. Loss of A20 and BTK C481F mutation as mechanisms of BTK inhibitor resistance in TMD8.
(A) Cell viability of BTK inhibitor (BTKi)-sensitive (TMD8BTKi-S) and BTKi-resistant (TMD8BTKi-R) cells to ibrutinib, 96 hour CellTiterGlo assay, mean ± SEM, n = 4. (B) Protein lysates were generated for TMD8 (DMS0 control), TMD8A20-Q143*and TMD8BTK-C481F cell lines, and protein expression of A20, p-IκBα, total IκBα, p-BTK and total BTK was analyzed using Simple Western. (C) Cell viability of BTKi-resistant TMD8A20-Q143* (left) or BTKi-resistant TMD8BTK-C481F (right) treated with ONO/GS-4059, idelalisib, or a combination of idelalisib plus ONO/GS-4059 (50 nM), mean ± SEM, n = 4. (D) Lysates of TMD8 DMSO control and TMD8A20-Q143* lines were analyzed by western blot (p-ERK 1/2, p-AKT S473, AKT) and Simple Western (p-BTK, p-IκBα S32, IκBα). Cells were treated with idelalisib (420 nM), ONO/GS-4059 (320 nM) or in combination for 24 hours. (E) Quantification of p-ERK1/2 and p-IκBα S32 blot results from Fig 5D. Values are normalized to actin and graphed as percent of DMSO vehicle control. Simple Western and western blot images are representative experiments from n = 2–3 experiments.

Fig 6. Schematic summarizing the proposed mechanisms…

Fig 6. Schematic summarizing the proposed mechanisms of PI3Kδ and BTK inhibitor resistance.

Upon antigen…

Fig 6. Schematic summarizing the proposed mechanisms of PI3Kδ and BTK inhibitor resistance.
Upon antigen binding to the B-cell receptor (BCR), the PI3K, MAPK and NF-kB pathways are activated through a cascade of phosphorylation events. BCR stimulation recruits SYK to the membrane, where it is phosphorylated and then subsequently phosphorylates BTK. BTK activation leads to subsequent dual phosphorylation and activation of PLCγ2, which leads to downstream generation of the second messengers IP and Dag. Active PI3Kδ is recruited to the membrane and converts PIP2 to PIP3, leading to subsequent activation and phosphorylation of PDK1 and AKT. PTEN is a phosphatase which negatively regulates the PI3K pathway. PKC can be activated downstream of both PI3K and BTK leading to NF-kB and MAPK pathway activation [27]. In our idelalisib resistance model, PTEN is lost which results in the hyperactivation of the PI3K pathway. Resistance can be overcome by inhibiting PI3Kδ and the downstream phosphoproteins PDK1 and AKT. In the BTK resistance model, A20 is lost which leads to hyperactivation of the NF-κB pathway which could be overcome by inhibiting BTK and PI3Kδ. Additionally, the BTK inhibitor binding site at C481 was mutated resulting in ONO/GS-4059 resistance.
Fig 3. Combination of idelalisib and ONO/GS-4059…
Fig 3. Combination of idelalisib and ONO/GS-4059 can overcome TMD8 acquired idelalisib resistance.
(A) Cell viability of vehicle control line (TMD8IDELA-S) or idelalisib resistant line (TMD8IDELA-R) in response to idelalisib treatment, 96 hour CellTiterGlo assay, mean ± SEM, n = 4. (B) Cell viability of TMD8IDELA-S (top graph) or TMD8IDELA-R cells (bottom graph) treated with idelalisib, ONO/GS-4059 or ONO/GS-4059 in combination with idelalisib (1 μM), n = 4, mean ± SEM. Combination curve is normalized to the single agent alone. (C) Protein lysates from TMD8IDELA-S (vehicle treated) and TMD8IDELA-R treated with vehicle, idelalisib (420 nM), ONO/GS-4059 (320 nM) or in combination for 2 hours were generated and analyzed in a single run using Simple Western, representative image of a single run of n = 3. (D) Quantification of Fig 3C showing AUC normalization to actin, and values normalized to TMD8IDELA-S DMSO treated control for all treatment groups from a single run.
Fig 4. PI3K upregulation in acquired resistance…
Fig 4. PI3K upregulation in acquired resistance is sensitive to idelalisib and AKT or PDK1 inhibitor combinations.
(A) Protein lysates were generated from TMD8IDELA-S and TMD8IDELA-R cells, and analyzed by western blot (p-AKT S473, p-AKT T308, AKT, p-S6RP S235/236, S6RP, p-GSK3β S9, GSK3β) and Simple Western (PTEN). (B) Cell viability of TMD8IDELA-S (left) and TMD8IDELA-R (right) cells treated with idelalisib, MK-2206 or combination of idelalisib and MK-2206 (1 μM), 96 hours CellTiterGlo assay, mean ± SEM, n = 4. Combination curve is normalized to the single agent alone. (C) Cell viability of TMD8IDELA-S (left) and TMD8IDELA-R (right) cells treated with idelalisib, GSK2334470 or combination of idelalisib and GSK2334470 (3 μM), 96 hours CellTiterGlo assay, mean ± SEM, n = 4. Combination curve is normalized to the single agent alone. (D) TMD8IDELA-R cells were treated with vehicle, idelalisib (1 μM), MK-2206 (1 μM), and idelalisib plus MK-2206, or (E) Vehicle, idelalisib (1 μM), GSK2334470 (1 μM), and idelalisib plus GSK2334470 for 48 hours. Apoptosis was assessed by 7AAD and PE annexin V staining, and analyzed by flow cytometry, mean ± SD, n = 3. (F) TMD8IDELA-R cells were treated with vehicle, idelalisib (1 μM), MK-2206 (1 μM), or idelalisib plus MK-2206 and (G) Vehicle, idelalisib (1 μM), GSK2334470 (1 μM), or idelalisib plus GSK2334470 for 2 hours. Protein lysates were generated and analyzed by western blot, representative experiment of n = 3. TMD8IDELA-S vehicle control was included as a reference on the same membrane.
Fig 5. Loss of A20 and BTK…
Fig 5. Loss of A20 and BTK C481F mutation as mechanisms of BTK inhibitor resistance in TMD8.
(A) Cell viability of BTK inhibitor (BTKi)-sensitive (TMD8BTKi-S) and BTKi-resistant (TMD8BTKi-R) cells to ibrutinib, 96 hour CellTiterGlo assay, mean ± SEM, n = 4. (B) Protein lysates were generated for TMD8 (DMS0 control), TMD8A20-Q143*and TMD8BTK-C481F cell lines, and protein expression of A20, p-IκBα, total IκBα, p-BTK and total BTK was analyzed using Simple Western. (C) Cell viability of BTKi-resistant TMD8A20-Q143* (left) or BTKi-resistant TMD8BTK-C481F (right) treated with ONO/GS-4059, idelalisib, or a combination of idelalisib plus ONO/GS-4059 (50 nM), mean ± SEM, n = 4. (D) Lysates of TMD8 DMSO control and TMD8A20-Q143* lines were analyzed by western blot (p-ERK 1/2, p-AKT S473, AKT) and Simple Western (p-BTK, p-IκBα S32, IκBα). Cells were treated with idelalisib (420 nM), ONO/GS-4059 (320 nM) or in combination for 24 hours. (E) Quantification of p-ERK1/2 and p-IκBα S32 blot results from Fig 5D. Values are normalized to actin and graphed as percent of DMSO vehicle control. Simple Western and western blot images are representative experiments from n = 2–3 experiments.
Fig 6. Schematic summarizing the proposed mechanisms…
Fig 6. Schematic summarizing the proposed mechanisms of PI3Kδ and BTK inhibitor resistance.
Upon antigen binding to the B-cell receptor (BCR), the PI3K, MAPK and NF-kB pathways are activated through a cascade of phosphorylation events. BCR stimulation recruits SYK to the membrane, where it is phosphorylated and then subsequently phosphorylates BTK. BTK activation leads to subsequent dual phosphorylation and activation of PLCγ2, which leads to downstream generation of the second messengers IP and Dag. Active PI3Kδ is recruited to the membrane and converts PIP2 to PIP3, leading to subsequent activation and phosphorylation of PDK1 and AKT. PTEN is a phosphatase which negatively regulates the PI3K pathway. PKC can be activated downstream of both PI3K and BTK leading to NF-kB and MAPK pathway activation [27]. In our idelalisib resistance model, PTEN is lost which results in the hyperactivation of the PI3K pathway. Resistance can be overcome by inhibiting PI3Kδ and the downstream phosphoproteins PDK1 and AKT. In the BTK resistance model, A20 is lost which leads to hyperactivation of the NF-κB pathway which could be overcome by inhibiting BTK and PI3Kδ. Additionally, the BTK inhibitor binding site at C481 was mutated resulting in ONO/GS-4059 resistance.

References

    1. Jones JA, Byrd JC. How will B-cell-receptor–targeted therapies change future CLL therapy? Blood. 2014;123:1455–1460. 10.1182/blood-2013-09-453092
    1. Woyach JA, Johnson AJ, Byrd JC. The B-cell receptor signaling pathway as a therapeutic target in CLL. Blood. 2012;120:1175–1184. 10.1182/blood-2012-02-362624
    1. Niemann CU, Wiestner A. B-cell receptor signaling as a driver of lymphoma development and evolution. Semin Cancer Biol. 2013;23:410–421. 10.1016/j.semcancer.2013.09.001
    1. Vanhaesebroeck B, Whitehead MA, Pineiro R. Molecules in medicine mini-review: isoforms of PI3K in biology and disease. J Mol Med (Berl). 2016;94:5–11.
    1. Brown JR. The PI3K pathway: clinical inhibition in chronic lymphocytic leukemia. Semin Oncol. 2016;43:260–264. 10.1053/j.seminoncol.2016.02.004
    1. Graf SA, Gopal AK. Idelalisib for the treatment of non-Hodgkin lymphoma. Expert Opin Pharmacother. 2016;17:265–274. 10.1517/14656566.2016.1135130
    1. US Food and Drug Administration. FDA expands approved use of Imbruvica for chronic lymphocytic leukemia. FDA News Release. . Accessed March 25, 2016. 2014.
    1. US Food and Drug Administration. FDA approves Zydelig for three types of blood cancers. FDA News Release. . Accessed March 25, 2016. 2014.
    1. Yang Q, Modi P, Newcomb T, Quéva C, Gandhi V. Idelalisib: First-in-class PI3K delta inhibitor for the treatment of chronic lymphocytic leukemia, small lymphocytic leukemia, and follicular lymphoma. Clin Cancer Res. 2015;21:1537–1542. 10.1158/1078-0432.CCR-14-2034
    1. Walter HS, Rule SA, Dyer MJ, Karlin L, Jones C, Cazin B, et al. A phase 1 clinical trial of the selective BTK inhibitor ONO/GS-4059 in relapsed and refractory mature B-cell malignancies. Blood. 2016;127:411–419. 10.1182/blood-2015-08-664086
    1. Patel L, Chandrasekhar J, Evarts J, Forseth K, Haran AC, Ip C, et al. Discovery of orally efficacious phosphoinositide 3-kinase delta inhibitors with improved metabolic stability. J Med Chem. 2016.
    1. Fritsch C, Huang A, Chatenay-Rivauday C, Schnell C, Reddy A, Liu M, et al. Characterization of the novel and specific PI3Kalpha inhibitor NVP-BYL719 and development of the patient stratification strategy for clinical trials. Mol Cancer Ther. 2014;13:1117–1129. 10.1158/1535-7163.MCT-13-0865
    1. Nylander S, Kull B, Bjorkman JA, Ulvinge JC, Oakes N, Emanuelsson BM, et al. Human target validation of phosphoinositide 3-kinase (PI3K)beta: effects on platelets and insulin sensitivity, using AZD6482 a novel PI3K beta inhibitor. J Thromb Haemost. 2012;10:2127–2136. 10.1111/j.1538-7836.2012.04898.x
    1. Folkes AJ, Ahmadi K, Alderton WK, Alix S, Baker SJ, Box G, et al. The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-piperazin-1-ylmethyl)-4-morpholin-4-yl-t hieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer. J Med Chem. 2008;51:5522–5532. 10.1021/jm800295d
    1. Yap TA, Yan L, Patnaik A, Fearen I, Olmos D, Papadopoulos K, et al. First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors. J Clin Oncol. 2011;29:4688–4695. 10.1200/JCO.2011.35.5263
    1. Najafov A, Sommer EM, Axten JM, Deyoung MP, Alessi DR. Characterization of GSK2334470, a novel and highly specific inhibitor of PDK1. Biochem J. 2011;433:357–369. 10.1042/BJ20101732
    1. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10:R25 10.1186/gb-2009-10-3-r25
    1. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576. 10.1101/gr.129684.111
    1. Cingolani P, Platts A, Wang le L, Coon M, Nguyen T, Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012;6:80–92.
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. 10.1093/bioinformatics/bts635
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;12:323 10.1186/1471-2105-12-323
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. 10.1093/bioinformatics/btp616
    1. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47 10.1093/nar/gkv007
    1. Kim Y, Ju H, Kim DH, Yoo HY, Kim SJ, Kim WS, et al. CD79B and MYD88 mutations in diffuse large B-cell lymphoma. Human Pathology. 2014;45:556–564. 10.1016/j.humpath.2013.10.023.
    1. Somoza JR, Koditek D, Villasenor AG, Novikov N, Wong MH, Liclican A, et al. Structural, biochemical, and biophysical characterization of idelalisib binding to phosphoinositide 3-kinase delta. J Biol Chem. 2015;290:8439–8446. 10.1074/jbc.M114.634683
    1. Burger MT, Pecchi S, Wagman A, Ni ZJ, Knapp M, Hendrickson T, et al. Identification of NVP-BKM120 as a Potent, Selective, Orally Bioavailable Class I PI3 Kinase Inhibitor for Treating Cancer. ACS Med Chem Letters. 2011;2:774–779.
    1. Stevenson FK, Krysov S, Davies AJ, Steele AJ, Packham G. B-cell receptor signaling in chronic lymphocytic leukemia. Blood. 2011;118:4313–4320. 10.1182/blood-2011-06-338855
    1. Grammer TC, Blenis J. Evidence for MEK-independent pathways regulating the prolonged activation of the ERK-MAP kinases. Oncogene. 1997;14:1635–1642. 10.1038/sj.onc.1201000
    1. Nore BF, Vargas L, Mohamed AJ, Branden LJ, Backesjo CM, Islam TC, et al. Redistribution of Bruton's tyrosine kinase by activation of phosphatidylinositol 3-kinase and Rho-family GTPases. Eur J Immunol. 2000;30:145–154. 10.1002/1521-4141(200001)30:1<145::AID-IMMU145>;2-0
    1. Varnai P, Rother KI, Balla T. Phosphatidylinositol 3-kinase-dependent membrane association of the Bruton's tyrosine kinase pleckstrin homology domain visualized in single living cells. J Biol Chem. 1999;274:10983–10989.
    1. Albihn A, Johnsen JI, Henriksson MA. MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res. 2010;107:163–224. 10.1016/S0065-230X(10)07006-5
    1. Liu YJ, Kenney T, Butterworth L, Kashishian A, Meadows S, Yue P, et al. Idelalisib has activity at clinically achievable drug concentrations in a subset of ABC and GCB diffuse large B-cell lymphoma and transformed follicular lymphoma cell lines. Cancer Res 2015;75 (Suppl):Abstract 2673.
    1. Yeomans A, Thirdborough SM, Valle-Argos B, Linley A, Krysov S, Hidalgo MS, et al. Engagement of the B-cell receptor of chronic lymphocytic leukemia cells drives global and MYC-specific mRNA translation. Blood. 2016;127:449–457. 10.1182/blood-2015-07-660969
    1. Trabucco SE, Gerstein RM, Evens AM, Bradner JE, Shultz LD, Greiner DL, et al. Inhibition of bromodomain proteins for the treatment of human diffuse large B-cell lymphoma. Clin Cancer Res. 2015;21:113–122. 10.1158/1078-0432.CCR-13-3346
    1. Huw LY, O'Brien C, Pandita A, Mohan S, Spoerke JM, Lu S, et al. Acquired PIK3CA amplification causes resistance to selective phosphoinositide 3-kinase inhibitors in breast cancer. Oncogenesis. 2013;2.
    1. Nakanishi Y, Walter K, Spoerke JM, O'Brien C, Huw LY, Hampton GM, et al. Activating mutations in PIK3CB confer resistance to PI3K inhibition and define a novel oncogenic role for p110beta. Cancer Res. 2016;76:1193–1203. 10.1158/0008-5472.CAN-15-2201
    1. Schwartz S, Wongvipat J, Trigwell CB, Hancox U, Carver BS, Rodrik-Outmezguine V, et al. Feedback suppression of PI3Kalpha signaling in PTEN-mutated tumors is relieved by selective inhibition of PI3Kbeta. Cancer Cell. 2015;27:109–122. 10.1016/j.ccell.2014.11.008
    1. Juric D, Castel P, Griffith M, Griffith OL, Won HH, Ellis H, et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)K alpha inhibitor. Nature. 2015;518:240–U230. 10.1038/nature13948
    1. Crystal AS, Shaw AT, Sequist LV, Friboulet L, Niederst MJ, Lockerman EL, et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science. 2014;346:1480–1486. 10.1126/science.1254721
    1. Elkabets M, Pazarentzos E, Juric D, Sheng Q, Pelossof RA, Brook S, et al. AXL mediates resistance to PI3Kalpha inhibition by activating the EGFR/PKC/mTOR axis in head and neck and esophageal squamous cell carcinomas. Cancer Cell. 2015;27:533–546. 10.1016/j.ccell.2015.03.010
    1. Woyach JA, Furman RR, Liu TM, Ozer HG, Zapatka M, Ruppert AS, et al. Resistance mechanisms for the Bruton's tyrosine kinase inhibitor ibrutinib. N Eng J Med. 2014;370:2286–2294.
    1. Zhang SQ, Smith SM, Zhang SY, Lynn Wang Y. Mechanisms of ibrutinib resistance in chronic lymphocytic leukaemia and non-Hodgkin lymphoma. Br J Haematol. 2015;170:445–456. 10.1111/bjh.13427
    1. Liu TM, Woyach JA, Zhong YM, Lozanski A, Lozanski G, Dong S, et al. Hypermorphic mutation of phospholipase C, gamma 2 acquired in ibrutinib-resistant CLL confers BTK independency upon B-cell receptor activation. Blood. 2015;126:61–68. 10.1182/blood-2015-02-626846
    1. Wilson WH, Young RM, Schmitz R, Yang Y, Pittaluga S, Wright G, et al. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med. 2015;21:922–926. 10.1038/nm.3884
    1. Compagno M, Lim WK, Grunn A, Nandula SV, Brahmachary M, Shen Q, et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature. 2009;459:717–721. 10.1038/nature07968
    1. Bojarczuk K, Sasi BK, Gobessi S, Innocenti I, Pozzato G, Laurenti L, et al. BCR signaling inhibitors differ in their ability to overcome Mcl-1-mediated resistance of CLL B cells to ABT-199. Blood. 2016;127:3192–3201. 10.1182/blood-2015-10-675009
    1. Kuo HP, Crowley R, Xue L, Schweighofer KJ, Cheung LWK, Hsieh S, et al. Combination of ibrutinib and BCL-2 or SYK inhibitors in ibrutinib resistant ABC-subtype of diffuse large B-Cell lymphoma. Blood. 2014;124.
    1. Hamasy A, Wang Q, Blomberg KE, Mohammad DK, Yu L, Vihinen M, et al. Substitution scanning identifies a novel, catalytically active ibrutinib-resistant BTK cysteine 481 to threonine (C481T) variant. Leukemia. 2016.
    1. Anthony Mato C N, Paul M. Barr, Chaitra S. Ujjani, Brian T. Hill et al. Favorable outcomes in CLL pts with alternate kinase inhibitors following ibrutinib or idelalisib discontinuation: results from a large multi-center study. Blood (ASH Annual Meeting Abstracts). 2015;126:719.

Source: PubMed

3
Suscribir