Current Advances and Future Strategies for BCL-2 Inhibitors: Potent Weapons against Cancers
Abstract
:Simple Summary
Abstract
1. Introduction
Compounds | Targets | Status | Applicable Cancer Types | Refs. |
---|---|---|---|---|
ABT199 (venetoclax) | BCL-2 | FDA-approved | Hematologic cancers, solid tumors | [11,12,13,14] |
S55746 (BCL201) | BCL-2 | Phase 1 | Hematologic cancers | [15,16] |
APG-2575 (lisaftoclax) | BCL-2 | Phase 1/2 | Hematologic cancers | [17,18] |
G3139 (oblimersen) | BCL-2 | Phase 1/2/3 | Hematologic cancers, solid tumors | [19,20,21] |
AZD4320 | BCL-2 and BCL-XL | Preclinical | Hematologic cancers, malignant pleural mesothelioma | [22,23] |
AZD0466 | BCL-2 and BCL-XL | Phase 1/2 | Hematologic cancers, solid tumors | [23,24] |
APG-1252 (pelcitoclax) | BCL-2 and BCL-XL | Phase 1/2 | NSCLC, NPC, colorectal cancer, AML | [25,26,27,28] |
BM-1197 | BCL-2 and BCL-XL | Preclinical | SCLC, adenoid cystic carcinoma, NHL | [29,30,31] |
S44563 | BCL-2 and BCL-XL | Preclinical | SCLC, uveal melanoma | [32,33] |
ABT-737 | BCL-2, BCL-XL, and BCL-W | Preclinical | Hematologic cancers, solid tumors | [3,4,5,6] |
ABT-263 (navitoclax) | BCL-2, BCL-XL, and BCL-W | Phase 1/2 | Hematologic cancers, solid tumors | [7,8,9,10] |
GX15-070 (obatoclax) | BCL-2, BCL-XL, BCL-W, and MCL-1 | Phase 1/2 | Hematologic cancers, solid tumors | [34,35,36] |
AT-101 | BCL-2, BCL-XL, BCL-W, and MCL-1 | Phase 1/2 | Hematologic cancers, solid tumors | [37,38] |
2. Mechanisms of Drug Action
3. Substantial Progress in Hematologic Malignancies
3.1. Chronic Lymphocytic Leukemia
Disease | Indications | Combined Agents | Treatment Outcomes | Phase | Clinical Trials | Ref. |
---|---|---|---|---|---|---|
CLL | R/R CLL or SLL | None | ORR: 79%; CR: 20%; 15-month PFS: 69% | 1 | NCT01328626 | [12] |
R/R CLL with del 17p | None | ORR: 79%; 2-year PFS: 54% | 2 | NCT01889186 | [44] | |
R/R CLL | Rituximab | ORR: 86%; CR: 51%; uMRD: 57% | 1 | NCT01682616 | [45] | |
R/R CLL | Rituximab | 4-year PFS: 57%; 4-year OS: 85% | 3 | NCT02005471 (MURANO) | [47] | |
Previously untreated CLL | Obinutuzumab | 2-year PFS: 88% | 3 | NCT02242942 (CLL14) | [49] | |
R/R CLL | Ibrutinib | ORR: 89%; CR: 51%; uMRD: 53% | 2 | ISCRTN13751862 (CLARITY) | [51] | |
Previously untreated CLL | Ibrutinib | uMRD: 75% | 2 | NCT02910583 (CAPTIVATE) | [52] | |
R/R CLL | Obinutuzumab and ibrutinib | ORR: 92%; CR or CRi: 42% | 1 | NCT02427451 | [53] | |
AML | R/R AML or unfit for intensive chemotherapy | None | ORR: 19%; CR: 13% | 2 | NCT01994837 | [55] |
Previously untreated elderly AML patients ineligible for intensive chemotherapy | Azacytidine or decitabine | CR or CRi: 61% | 1 | NCT02203773 | [13] | |
ND intensive chemotherapy ineligible and R/R AML | Decitabine | ORR: 74% | 2 | NCT03404193 | [56] | |
Previously untreated elderly AML patients ineligible for intensive chemotherapy | Azacytidine | ORR: 37%; CR or CRi: 66%; median OS: 14.7 months | 1 | NCT02993523 (VIALE-A) | [57] | |
Previously untreated elderly AML patients ineligible for intensive chemotherapy | Cytarabine | CR or CRi: 54%; median OS: 10.1 months; median DOR: 8.1 months | 1/2 | NCT02287233 | [58] | |
ND-AML ineligible for intensive chemotherapy | Cytarabine | CR or CRi: 48%; median OS: 7.2 months | 3 | NCT03069352 | [59] | |
ND-AML | FLAG- IDA | ORR: 97%; CRc: 90%; uMRD: 96%; 1-year OS: 94% | 1/2 | NCT03214562 | [60] | |
R/R AML | ORR: 72%; CRc: 66%; uMRD: 69%; 1-year OS: 78% | |||||
ND-AML | Decitabine and FLT3 inhibitor | CRc: 92%; uMRD: 56% | 2 | NCT03404193 | [61] | |
R/R AML | CRc: 62%; uMRD: 63% | |||||
MM | R/R MM | None | ORR: 21%; VGPR: 15% | 1 | NCT01794520 | [62] |
R/R MM | Bortezomib and dexamethasone | ORR: 67%; VGPR: 42%; median DOR: 9.7 months | 1 | NCT01794507 | [63] | |
R/R MM | Bortezomib and dexamethasone | Median PFS: 22%; TE-SAEs: 48% | 3 | NCT02755597 (BELLINI) | [64] | |
R/R MM patients refractory to lenalidomide | Pomalidomide and dexamethasone | ORR: 53%; median DOR: 12.9 months; median PFS: 10.5 months | 2 | NCT03567616 | [65] | |
R/R MM | Carfilzomib and dexamethasone | ORR: 80%; CR: 41%; median PFS: 22.8 months | 2 | NCT02899052 | [66] | |
R/R MM with t(11;14) | Daratumumab and dexamethasone | ORR: 96%; 1.5-year PFS: 91% | 1 | NCT03314181 | [67] | |
R/R MM | Daratumumab, dexamethasone, and bortezomib | ORR: 92%; 1.5-year PFS: 67% | ||||
NHL | R/R MCL | None | ORR: 75%; median PFS: 14 months | 1 | NCT01328626 | [68] |
R/R FL | ORR: 38%; median PFS: 11 months | |||||
R/R DLBCL | ORR: 18%; median PFS: 1 month | |||||
MCL | Ibrutinib | CR: 42%; uMRD: 67% | 2 | NCT02471391 (AIM) | [69] | |
Relapsed MCL | Ibrutinib and obinutuzumab | CR: 67%; uMRD: 72%; 2-year PFS: 70% | 1/2 | NCT02558816 | [70] | |
Untreated MCL | CR: 87%; uMRD: 100%; 1-year PFS: 93% | |||||
B-cell NHL | R-/G-CHOP | ORR: 88% | 1 | NCT02055820 (CAVALLI) | [71] | |
Previously untreated DLBCL | R-CHOP | CR: 69% | 2 | NCT02055820 (CAVALLI) | [72] | |
R/R FL | Rituximab | CR: 17% | 2 | NCT02187861 (CONTRALTO) | [73] | |
Rituximab and bendamustine | CR: 75% | |||||
ALL | R/R ALL or LBL | Navitoclax | CR: 60% | 1 | NCT03181126 | [74] |
3.2. Acute Myeloid Leukemia
3.3. Multiple Myeloma
3.4. Other Hematologic Malignancies
4. Genetic Mutations Associated with Treatment Effectiveness
4.1. BCL-2 Mutations
4.2. Therapeutically Favorable Mutations
4.3. Therapeutically Adverse Mutations
5. Potential Applications in Solid Tumors
5.1. Breast Cancer
5.2. Lung Cancer
5.3. Pancreatic Cancer
5.4. Sarcoma
5.5. Other Solid Tumors
Cancers | Cell Types | Synergistic Drugs | Drug Attributes | Ref. |
---|---|---|---|---|
Breast cancer | ER(+) breast cancer cell lines, patient-derived organoid, patient-derived xenograft | Palbociclib | CDK4/6 inhibitor | [129] |
LM2-4, BT549, MDA-157 | Dasatinib | SRC inhibitor | [130] | |
SKBR3, MDAMB468, T47D, CAMA-1 | AZD1775 | WEE1 inhibitor | [135] | |
SUM149, BT474, xenografts | Neratinib | ERBB1/2/4 inhibitor | [136] | |
MCF7, BT549, MDAMB231 | GSIXII | Gamma-secretase inhibitor | [134] | |
Patient samples, TNBC cell lines, and xenografts | Metformin | AMPK activator | [132] | |
Lung cancer | NCI-H146, H1963, xenografts | ABBV-075 | BET inhibitor | [143] |
SCLC cell lines and xenografts | Dinaciclib | CDK9 inhibitor | [144] | |
H69 and H82 | KAN0441571C | ROR1 inhibitor | [145] | |
PC-9 and xenografts | Gefitinib | EGFR-tyrosine kinase inhibitor | [149] | |
H157, H460, H1299, xenografts | Decitabine | DNA methyltransferase inhibitor | [148] | |
DMS53, H460, xenografts | BKA-073 | BAK activator | [151] | |
Pancreatic cancer | MIAPacCa-2, SW1990, xenografts | Gemcitabine | Chemotherapeutics | [153] |
SUIT-2, MIAPaCa-2, BxPC-3 | Prexasertib | CHK1 inhibitor | [156] | |
S2013 and MIAPaCa-2 | Analog 24 | CDK5 inhibitor | [157] | |
Soft tissue sarcomas | STS cell lines and tumor-derived cells | Bortezomib | Proteasome inhibitor | [159] |
RD, TE381.T, RH30, primary-derived RMS cells | JNJ | Histone deacetylase inhibitor | [160] | |
Primary cells, ES cell lines, patient-derived and cell line-derived xenografts | Olaparib | PARP inhibitor | [162] | |
Colorectal cancer | RKO cell line and xenografts | LZT-106 | CDK9 inhibitor | [168] |
RKO | Birinapant/AT-406 | IAP antagonist | [166] | |
HT-29 and HCT-116 | Perifosine | AKT inhibitor | [171] | |
Hepatocellular carcinoma | HepG2, Hep3B, xenografts | Osimertinib | EGFR-tyrosine kinase inhibitor | [169] |
HepG2 | Curcumin | Plant polyphenol | [172] | |
HepG2 and SMMC-7721 | Norcantharidin | Herbal components | [173] |
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Ashkenazi, A.; Fairbrother, W.J.; Leverson, J.D.; Souers, A.J. From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat. Rev. Drug Discov. 2017, 16, 273–284. [Google Scholar] [CrossRef] [PubMed]
- Diepstraten, S.T.; Anderson, M.A.; Czabotar, P.E.; Lessene, G.; Strasser, A.; Kelly, G.L. The manipulation of apoptosis for cancer therapy using BH3-mimetic drugs. Nat. Rev. Cancer 2021, 22, 45–64. [Google Scholar] [CrossRef] [PubMed]
- Oltersdorf, T.; Elmore, S.W.; Shoemaker, A.R.; Armstrong, R.C.; Augeri, D.J.; Belli, B.A.; Bruncko, M.; Deckwerth, T.L.; Dinges, J.; Hajduk, P.J.; et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 2005, 435, 677–681. [Google Scholar] [CrossRef]
- Del Gaizo Moore, V.; Brown, J.R.; Certo, M.; Love, T.M.; Novina, C.D.; Letai, A. Chronic lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737. J. Clin. Investig. 2007, 117, 112–121. [Google Scholar] [CrossRef] [PubMed]
- Del Gaizo Moore, V.; Schlis, K.D.; Sallan, S.E.; Armstrong, S.A.; Letai, A. BCL-2 dependence and ABT-737 sensitivity in acute lymphoblastic leukemia. Blood 2008, 111, 2300–2309. [Google Scholar] [CrossRef]
- Hikita, H.; Takehara, T.; Shimizu, S.; Kodama, T.; Shigekawa, M.; Iwase, K.; Hosui, A.; Miyagi, T.; Tatsumi, T.; Ishida, H.; et al. The Bcl-xL inhibitor, ABT-737, efficiently induces apoptosis and suppresses growth of hepatoma cells in combination with sorafenib. Hepatology 2010, 52, 1310–1321. [Google Scholar] [CrossRef]
- Tse, C.; Shoemaker, A.R.; Adickes, J.; Anderson, M.G.; Chen, J.; Jin, S.; Johnson, E.F.; Marsh, K.C.; Mitten, M.J.; Nimmer, P.; et al. ABT-263: A potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008, 68, 3421–3428. [Google Scholar] [CrossRef]
- Wilson, W.H.; O’Connor, O.A.; Czuczman, M.S.; LaCasce, A.S.; Gerecitano, J.F.; Leonard, J.P.; Tulpule, A.; Dunleavy, K.; Xiong, H.; Chiu, Y.L.; et al. Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: A phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity. Lancet Oncol. 2010, 11, 1149–1159. [Google Scholar] [CrossRef]
- Roberts, A.W.; Seymour, J.F.; Brown, J.R.; Wierda, W.G.; Kipps, T.J.; Khaw, S.L.; Carney, D.A.; He, S.Z.; Huang, D.C.; Xiong, H.; et al. Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: Results of a phase I study of navitoclax in patients with relapsed or refractory disease. J. Clin. Oncol. 2012, 30, 488–496. [Google Scholar] [CrossRef]
- Nor Hisam, N.S.; Ugusman, A.; Rajab, N.F.; Ahmad, M.F.; Fenech, M.; Liew, S.L.; Mohamad Anuar, N.N. Combination Therapy of Navitoclax with Chemotherapeutic Agents in Solid Tumors and Blood Cancer: A Review of Current Evidence. Pharmaceutics 2021, 13, 1353. [Google Scholar] [CrossRef]
- Souers, A.J.; Leverson, J.D.; Boghaert, E.R.; Ackler, S.L.; Catron, N.D.; Chen, J.; Dayton, B.D.; Ding, H.; Enschede, S.H.; Fairbrother, W.J.; et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat. Med. 2013, 19, 202–208. [Google Scholar] [CrossRef] [PubMed]
- Roberts, A.W.; Davids, M.S.; Pagel, J.M.; Kahl, B.S.; Puvvada, S.D.; Gerecitano, J.F.; Kipps, T.J.; Anderson, M.A.; Brown, J.R.; Gressick, L.; et al. Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia. N. Engl. J. Med. 2016, 374, 311–322. [Google Scholar] [CrossRef] [PubMed]
- DiNardo, C.D.; Pratz, K.W.; Letai, A.; Jonas, B.A.; Wei, A.H.; Thirman, M.; Arellano, M.; Frattini, M.G.; Kantarjian, H.; Popovic, R.; et al. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: A non-randomised, open-label, phase 1b study. Lancet Oncol. 2018, 19, 216–228. [Google Scholar] [CrossRef]
- Lok, S.W.; Whittle, J.R.; Vaillant, F.; Teh, C.E.; Lo, L.L.; Policheni, A.N.; Bergin, A.R.T.; Desai, J.; Ftouni, S.; Gandolfo, L.C.; et al. A Phase Ib Dose-Escalation and Expansion Study of the BCL2 Inhibitor Venetoclax Combined with Tamoxifen in ER and BCL2-Positive Metastatic Breast Cancer. Cancer Discov. 2019, 9, 354–369. [Google Scholar] [CrossRef] [PubMed]
- Casara, P.; Davidson, J.; Claperon, A.; Le Toumelin-Braizat, G.; Vogler, M.; Bruno, A.; Chanrion, M.; Lysiak-Auvity, G.; Le Diguarher, T.; Starck, J.B.; et al. S55746 is a novel orally active BCL-2 selective and potent inhibitor that impairs hematological tumor growth. Oncotarget 2018, 9, 20075–20088. [Google Scholar] [CrossRef]
- Moujalled, D.M.; Pomilio, G.; Ghiurau, C.; Ivey, A.; Salmon, J.; Rijal, S.; Macraild, S.; Zhang, L.; Teh, T.C.; Tiong, I.S.; et al. Combining BH3-mimetics to target both BCL-2 and MCL1 has potent activity in pre-clinical models of acute myeloid leukemia. Leukemia 2019, 33, 905–917. [Google Scholar] [CrossRef]
- Deng, J.; Paulus, A.; Fang, D.D.; Manna, A.; Wang, G.; Wang, H.; Zhu, S.; Chen, J.; Min, P.; Yin, Y.; et al. Lisaftoclax (APG-2575) Is a Novel BCL-2 Inhibitor with Robust Antitumor Activity in Preclinical Models of Hematologic Malignancy. Clin. Cancer Res. 2022, 28, 5455–5468. [Google Scholar] [CrossRef]
- Ailawadhi, S.; Chen, Z.; Huang, B.; Paulus, A.; Collins, M.C.; Fu, L.T.; Li, M.; Ahmad, M.; Men, L.; Wang, H.; et al. Novel BCL-2 Inhibitor Lisaftoclax in Relapsed or Refractory Chronic Lymphocytic Leukemia and Other Hematologic Malignancies: First-in-Human Open-Label Trial. Clin. Cancer Res. 2023, 29, 2385–2393. [Google Scholar] [CrossRef] [PubMed]
- Marshall, J.; Chen, H.; Yang, D.; Figueira, M.; Bouker, K.B.; Ling, Y.; Lippman, M.; Frankel, S.R.; Hayes, D.F. A phase I trial of a Bcl-2 antisense (G3139) and weekly docetaxel in patients with advanced breast cancer and other solid tumors. Ann. Oncol. 2004, 15, 1274–1283. [Google Scholar] [CrossRef] [PubMed]
- O’Brien, S.; Moore, J.O.; Boyd, T.E.; Larratt, L.M.; Skotnicki, A.B.; Koziner, B.; Chanan-Khan, A.A.; Seymour, J.F.; Gribben, J.; Itri, L.M.; et al. 5-year survival in patients with relapsed or refractory chronic lymphocytic leukemia in a randomized, phase III trial of fludarabine plus cyclophosphamide with or without oblimersen. J. Clin. Oncol. 2009, 27, 5208–5212. [Google Scholar] [CrossRef]
- Walker, A.R.; Marcucci, G.; Yin, J.; Blum, W.; Stock, W.; Kohlschmidt, J.; Mrozek, K.; Carroll, A.J.; Eisfeld, A.K.; Wang, E.S.; et al. Phase 3 randomized trial of chemotherapy with or without oblimersen in older AML patients: CALGB 10201 (Alliance). Blood Adv. 2021, 5, 2775–2787. [Google Scholar] [CrossRef]
- Balachander, S.B.; Criscione, S.W.; Byth, K.F.; Cidado, J.; Adam, A.; Lewis, P.; Macintyre, T.; Wen, S.; Lawson, D.; Burke, K.; et al. AZD4320, A Dual Inhibitor of Bcl-2 and Bcl-x(L), Induces Tumor Regression in Hematologic Cancer Models without Dose-limiting Thrombocytopenia. Clin. Cancer Res. 2020, 26, 6535–6549. [Google Scholar] [CrossRef]
- Arulananda, S.; O’Brien, M.; Evangelista, M.; Jenkins, L.J.; Poh, A.R.; Walkiewicz, M.; Leong, T.; Mariadason, J.M.; Cebon, J.; Balachander, S.B.; et al. A novel BH3-mimetic, AZD0466, targeting BCL-XL and BCL-2 is effective in pre-clinical models of malignant pleural mesothelioma. Cell Death Discov. 2021, 7, 122. [Google Scholar] [CrossRef]
- Patterson, C.M.; Balachander, S.B.; Grant, I.; Pop-Damkov, P.; Kelly, B.; McCoull, W.; Parker, J.; Giannis, M.; Hill, K.J.; Gibbons, F.D.; et al. Design and optimisation of dendrimer-conjugated Bcl-2/x(L) inhibitor, AZD0466, with improved therapeutic index for cancer therapy. Commun. Biol. 2021, 4, 112. [Google Scholar] [CrossRef]
- Wang, J.; Yang, D.; Luo, Q.; Qiu, M.; Zhang, L.; Li, B.; Chen, H.; Yi, H.; Yan, X.; Li, S.; et al. APG-1252-12A induces mitochondria-dependent apoptosis through inhibiting the antiapoptotic proteins Bcl-2/Bcl-xl in HL-60 cells. Int. J. Oncol. 2017, 51, 563–572. [Google Scholar] [CrossRef]
- Luo, F.; Lu, F.T.; Qiu, M.Z.; Zhou, T.; Ma, W.J.; Luo, M.; Zeng, K.M.; Luo, Q.Y.; Pan, W.T.; Zhang, L.; et al. Gemcitabine and APG-1252, a novel small molecule inhibitor of BCL-2/BCL-XL, display a synergistic antitumor effect in nasopharyngeal carcinoma through the JAK-2/STAT3/MCL-1 signaling pathway. Cell Death Dis. 2021, 12, 772. [Google Scholar] [CrossRef] [PubMed]
- Qian, L.; Vallega, K.A.; Yao, W.; Wang, D.; Zhai, Y.; He, X.; Sun, S.Y. Therapeutic potential of the novel Bcl-2/Bcl-X(L) dual inhibitor, APG1252, alone or in combination against non-small cell lung cancer. Mol. Carcinog. 2022, 61, 1031–1042. [Google Scholar] [CrossRef]
- Yao, W.; Bai, L.; Wang, S.; Zhai, Y.; Sun, S.Y. Mcl-1 levels critically impact the sensitivities of human colorectal cancer cells to APG-1252-M1, a novel Bcl-2/Bcl-X(L) dual inhibitor that induces Bax-dependent apoptosis. Neoplasia 2022, 29, 100798. [Google Scholar] [CrossRef]
- Bai, L.; Chen, J.; McEachern, D.; Liu, L.; Zhou, H.; Aguilar, A.; Wang, S. BM-1197: A novel and specific Bcl-2/Bcl-xL inhibitor inducing complete and long-lasting tumor regression in vivo. PLoS ONE 2014, 9, e99404. [Google Scholar] [CrossRef] [PubMed]
- Acasigua, G.A.; Warner, K.A.; Nor, F.; Helman, J.; Pearson, A.T.; Fossati, A.C.; Wang, S.; Nor, J.E. BH3-mimetic small molecule inhibits the growth and recurrence of adenoid cystic carcinoma. Oral Oncol. 2015, 51, 839–847. [Google Scholar] [CrossRef]
- Sun, Y.L.; Jiang, W.Q.; Luo, Q.Y.; Yang, D.J.; Cai, Y.C.; Huang, H.Q.; Sun, J. A novel Bcl-2 inhibitor, BM-1197, induces apoptosis in malignant lymphoma cells through the endogenous apoptotic pathway. BMC Cancer 2019, 20, 1. [Google Scholar] [CrossRef]
- Loriot, Y.; Mordant, P.; Dugue, D.; Geneste, O.; Gombos, A.; Opolon, P.; Guegan, J.; Perfettini, J.L.; Pierre, A.; Berthier, L.K.; et al. Radiosensitization by a novel Bcl-2 and Bcl-XL inhibitor S44563 in small-cell lung cancer. Cell Death Dis. 2014, 5, e1423. [Google Scholar] [CrossRef]
- Nemati, F.; de Montrion, C.; Lang, G.; Kraus-Berthier, L.; Carita, G.; Sastre-Garau, X.; Berniard, A.; Vallerand, D.; Geneste, O.; de Plater, L.; et al. Targeting Bcl-2/Bcl-XL induces antitumor activity in uveal melanoma patient-derived xenografts. PLoS ONE 2014, 9, e80836. [Google Scholar] [CrossRef]
- Trudel, S.; Li, Z.H.; Rauw, J.; Tiedemann, R.E.; Wen, X.Y.; Stewart, A.K. Preclinical studies of the pan-Bcl inhibitor obatoclax (GX015-070) in multiple myeloma. Blood 2007, 109, 5430–5438. [Google Scholar] [CrossRef]
- Hwang, J.J.; Kuruvilla, J.; Mendelson, D.; Pishvaian, M.J.; Deeken, J.F.; Siu, L.L.; Berger, M.S.; Viallet, J.; Marshall, J.L. Phase I dose finding studies of obatoclax (GX15-070), a small molecule pan-BCL-2 family antagonist, in patients with advanced solid tumors or lymphoma. Clin. Cancer Res. 2010, 16, 4038–4045. [Google Scholar] [CrossRef]
- Rahmani, M.; Aust, M.M.; Attkisson, E.; Williams, D.C., Jr.; Ferreira-Gonzalez, A.; Grant, S. Inhibition of Bcl-2 antiapoptotic members by obatoclax potently enhances sorafenib-induced apoptosis in human myeloid leukemia cells through a Bim-dependent process. Blood 2012, 119, 6089–6098. [Google Scholar] [CrossRef]
- Yang, Q.; Chen, K.; Zhang, L.; Feng, L.; Fu, G.; Jiang, S.; Bi, S.; Lin, C.; Zhou, Y.; Zhao, H.; et al. Synthetic lethality of combined AT-101 with idarubicin in acute myeloid leukemia via blockade of DNA repair and activation of intrinsic apoptotic pathway. Cancer Lett. 2019, 461, 31–43. [Google Scholar] [CrossRef]
- Renner, O.; Mayer, M.; Leischner, C.; Burkard, M.; Berger, A.; Lauer, U.M.; Venturelli, S.; Bischoff, S.C. Systematic Review of Gossypol/AT-101 in Cancer Clinical Trials. Pharmaceuticals 2022, 15, 144. [Google Scholar] [CrossRef]
- Wong, R.S. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef]
- Singh, R.; Letai, A.; Sarosiek, K. Regulation of apoptosis in health and disease: The balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 2019, 20, 175–193. [Google Scholar] [CrossRef]
- Kale, J.; Osterlund, E.J.; Andrews, D.W. BCL-2 family proteins: Changing partners in the dance towards death. Cell Death Differ. 2018, 25, 65–80. [Google Scholar] [CrossRef] [PubMed]
- Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef]
- Roberts, A.W.; Wei, A.H.; Huang, D.C.S. BCL2 and MCL1 inhibitors for hematologic malignancies. Blood 2021, 138, 1120–1136. [Google Scholar] [CrossRef]
- Stilgenbauer, S.; Eichhorst, B.; Schetelig, J.; Coutre, S.; Seymour, J.F.; Munir, T.; Puvvada, S.D.; Wendtner, C.M.; Roberts, A.W.; Jurczak, W.; et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: A multicentre, open-label, phase 2 study. Lancet Oncol. 2016, 17, 768–778. [Google Scholar] [CrossRef]
- Seymour, J.F.; Ma, S.; Brander, D.M.; Choi, M.Y.; Barrientos, J.; Davids, M.S.; Anderson, M.A.; Beaven, A.W.; Rosen, S.T.; Tam, C.S.; et al. Venetoclax plus rituximab in relapsed or refractory chronic lymphocytic leukaemia: A phase 1b study. Lancet Oncol. 2017, 18, 230–240. [Google Scholar] [CrossRef] [PubMed]
- Kater, A.P.; Seymour, J.F.; Hillmen, P.; Eichhorst, B.; Langerak, A.W.; Owen, C.; Verdugo, M.; Wu, J.; Punnoose, E.A.; Jiang, Y.; et al. Fixed Duration of Venetoclax-Rituximab in Relapsed/Refractory Chronic Lymphocytic Leukemia Eradicates Minimal Residual Disease and Prolongs Survival: Post-Treatment Follow-Up of the MURANO Phase III Study. J. Clin. Oncol. 2019, 37, 269–277. [Google Scholar] [CrossRef]
- Kater, A.P.; Wu, J.Q.; Kipps, T.; Eichhorst, B.; Hillmen, P.; D’Rozario, J.; Assouline, S.; Owen, C.; Robak, T.; de la Serna, J.; et al. Venetoclax Plus Rituximab in Relapsed Chronic Lymphocytic Leukemia: 4-Year Results and Evaluation of Impact of Genomic Complexity and Gene Mutations From the MURANO Phase III Study. J. Clin. Oncol. 2020, 38, 4042–4054. [Google Scholar] [CrossRef]
- Ma, S.; Seymour, J.F.; Brander, D.M.; Kipps, T.J.; Choi, M.Y.; Anderson, M.A.; Humphrey, K.; Al Masud, A.; Pesko, J.; Nandam, R.; et al. Efficacy of venetoclax plus rituximab for relapsed CLL: 5-year follow-up of continuous or limited- duration therapy. Blood 2021, 138, 836–846. [Google Scholar] [CrossRef] [PubMed]
- Fischer, K.; Al-Sawaf, O.; Bahlo, J.; Fink, A.M.; Tandon, M.; Dixon, M.; Robrecht, S.; Warburton, S.; Humphrey, K.; Samoylova, O.; et al. Venetoclax and Obinutuzumab in Patients with CLL and Coexisting Conditions. N. Engl. J. Med. 2019, 380, 2225–2236. [Google Scholar] [CrossRef]
- Al-Sawaf, O.; Zhang, C.; Tandon, M.; Sinha, A.; Fink, A.M.; Robrecht, S.; Samoylova, O.; Liberati, A.M.; Pinilla-Ibarz, J.; Opat, S.; et al. Venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab for previously untreated chronic lymphocytic leukaemia (CLL14): Follow-up results from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2020, 21, 1188–1200. [Google Scholar] [CrossRef]
- Hillmen, P.; Rawstron, A.C.; Brock, K.; Munoz-Vicente, S.; Yates, F.J.; Bishop, R.; Boucher, R.; MacDonald, D.; Fegan, C.; McCaig, A.; et al. Ibrutinib Plus Venetoclax in Relapsed/Refractory Chronic Lymphocytic Leukemia: The CLARITY Study. J. Clin. Oncol. 2019, 37, 2722–2729. [Google Scholar] [CrossRef]
- Wierda, W.G.; Allan, J.N.; Siddiqi, T.; Kipps, T.J.; Opat, S.; Tedeschi, A.; Badoux, X.C.; Kuss, B.J.; Jackson, S.; Moreno, C.; et al. Ibrutinib Plus Venetoclax for First-Line Treatment of Chronic Lymphocytic Leukemia: Primary Analysis Results From the Minimal Residual Disease Cohort of the Randomized Phase II CAPTIVATE Study. J. Clin. Oncol. 2021, 39, 3853–3865. [Google Scholar] [CrossRef] [PubMed]
- Rogers, K.A.; Huang, Y.; Ruppert, A.S.; Awan, F.T.; Heerema, N.A.; Hoffman, C.; Lozanski, G.; Maddocks, K.J.; Moran, M.E.; Reid, M.A.; et al. Phase 1b study of obinutuzumab, ibrutinib, and venetoclax in relapsed and refractory chronic lymphocytic leukemia. Blood 2018, 132, 1568–1572. [Google Scholar] [CrossRef] [PubMed]
- Tambaro, F.P.; Wierda, W.G. Tumour lysis syndrome in patients with chronic lymphocytic leukaemia treated with BCL-2 inhibitors: Risk factors, prophylaxis, and treatment recommendations. Lancet Haematol. 2020, 7, e168–e176. [Google Scholar] [CrossRef]
- Konopleva, M.; Pollyea, D.A.; Potluri, J.; Chyla, B.; Hogdal, L.; Busman, T.; McKeegan, E.; Salem, A.H.; Zhu, M.; Ricker, J.L.; et al. Efficacy and Biological Correlates of Response in a Phase II Study of Venetoclax Monotherapy in Patients with Acute Myelogenous Leukemia. Cancer Discov. 2016, 6, 1106–1117. [Google Scholar] [CrossRef] [PubMed]
- DiNardo, C.D.; Maiti, A.; Rausch, C.R.; Pemmaraju, N.; Naqvi, K.; Daver, N.G.; Kadia, T.M.; Borthakur, G.; Ohanian, M.; Alvarado, Y.; et al. 10-day decitabine with venetoclax for newly diagnosed intensive chemotherapy ineligible, and relapsed or refractory acute myeloid leukaemia: A single-centre, phase 2 trial. Lancet Haematol. 2020, 7, e724–e736. [Google Scholar] [CrossRef] [PubMed]
- DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Dohner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef] [PubMed]
- Wei, A.H.; Strickland, S.A., Jr.; Hou, J.Z.; Fiedler, W.; Lin, T.L.; Walter, R.B.; Enjeti, A.; Tiong, I.S.; Savona, M.; Lee, S.; et al. Venetoclax Combined with Low-Dose Cytarabine for Previously Untreated Patients with Acute Myeloid Leukemia: Results From a Phase Ib/II Study. J. Clin. Oncol. 2019, 37, 1277–1284. [Google Scholar] [CrossRef]
- Wei, A.H.; Montesinos, P.; Ivanov, V.; DiNardo, C.D.; Novak, J.; Laribi, K.; Kim, I.; Stevens, D.A.; Fiedler, W.; Pagoni, M.; et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: A phase 3 randomized placebo-controlled trial. Blood 2020, 135, 2137–2145. [Google Scholar] [CrossRef]
- DiNardo, C.D.; Lachowiez, C.A.; Takahashi, K.; Loghavi, S.; Xiao, L.; Kadia, T.; Daver, N.; Adeoti, M.; Short, N.J.; Sasaki, K.; et al. Venetoclax Combined with FLAG-IDA Induction and Consolidation in Newly Diagnosed and Relapsed or Refractory Acute Myeloid Leukemia. J. Clin. Oncol. 2021, 39, 2768–2778. [Google Scholar] [CrossRef]
- Maiti, A.; DiNardo, C.D.; Daver, N.G.; Rausch, C.R.; Ravandi, F.; Kadia, T.M.; Pemmaraju, N.; Borthakur, G.; Bose, P.; Issa, G.C.; et al. Triplet therapy with venetoclax, FLT3 inhibitor and decitabine for FLT3-mutated acute myeloid leukemia. Blood Cancer J. 2021, 11, 25. [Google Scholar] [CrossRef]
- Kumar, S.; Kaufman, J.L.; Gasparetto, C.; Mikhael, J.; Vij, R.; Pegourie, B.; Benboubker, L.; Facon, T.; Amiot, M.; Moreau, P.; et al. Efficacy of venetoclax as targeted therapy for relapsed/refractory t(11;14) multiple myeloma. Blood 2017, 130, 2401–2409. [Google Scholar] [CrossRef]
- Moreau, P.; Chanan-Khan, A.; Roberts, A.W.; Agarwal, A.B.; Facon, T.; Kumar, S.; Touzeau, C.; Punnoose, E.A.; Cordero, J.; Munasinghe, W.; et al. Promising efficacy and acceptable safety of venetoclax plus bortezomib and dexamethasone in relapsed/refractory MM. Blood 2017, 130, 2392–2400. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.K.; Harrison, S.J.; Cavo, M.; de la Rubia, J.; Popat, R.; Gasparetto, C.; Hungria, V.; Salwender, H.; Suzuki, K.; Kim, I.; et al. Venetoclax or placebo in combination with bortezomib and dexamethasone in patients with relapsed or refractory multiple myeloma (BELLINI): A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2020, 21, 1630–1642. [Google Scholar] [CrossRef] [PubMed]
- Gasparetto, C.; Bowles, K.M.; Abdallah, A.O.; Morris, L.; Mander, G.; Coppola, S.; Wang, J.; Ross, J.A.; Bueno, O.F.; Arriola, E.; et al. A Phase II Study of Venetoclax in Combination with Pomalidomide and Dexamethasone in Relapsed/Refractory Multiple Myeloma. Clin. Lymphoma Myeloma Leuk. 2021, 21, 775–784. [Google Scholar] [CrossRef] [PubMed]
- Costa, L.J.; Davies, F.E.; Monohan, G.P.; Kovacsovics, T.; Burwick, N.; Jakubowiak, A.; Kaufman, J.L.; Hong, W.J.; Dail, M.; Salem, A.H.; et al. Phase 2 study of venetoclax plus carfilzomib and dexamethasone in patients with relapsed/refractory multiple myeloma. Blood Adv. 2021, 5, 3748–3759. [Google Scholar] [CrossRef]
- Bahlis, N.J.; Baz, R.; Harrison, S.J.; Quach, H.; Ho, S.J.; Vangsted, A.J.; Plesner, T.; Moreau, P.; Gibbs, S.D.; Coppola, S.; et al. Phase I Study of Venetoclax Plus Daratumumab and Dexamethasone, with or without Bortezomib, in Patients with Relapsed or Refractory Multiple Myeloma with and without t(11;14). J. Clin. Oncol. 2021, 39, 3602–3612. [Google Scholar] [CrossRef]
- Davids, M.S.; Roberts, A.W.; Seymour, J.F.; Pagel, J.M.; Kahl, B.S.; Wierda, W.G.; Puvvada, S.; Kipps, T.J.; Anderson, M.A.; Salem, A.H.; et al. Phase I First-in-Human Study of Venetoclax in Patients with Relapsed or Refractory Non-Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 826–833. [Google Scholar] [CrossRef] [PubMed]
- Tam, C.S.; Anderson, M.A.; Pott, C.; Agarwal, R.; Handunnetti, S.; Hicks, R.J.; Burbury, K.; Turner, G.; Di Iulio, J.; Bressel, M.; et al. Ibrutinib plus Venetoclax for the Treatment of Mantle-Cell Lymphoma. N. Engl. J. Med. 2018, 378, 1211–1223. [Google Scholar] [CrossRef] [PubMed]
- Le Gouill, S.; Morschhauser, F.; Chiron, D.; Bouabdallah, K.; Cartron, G.; Casasnovas, O.; Bodet-Milin, C.; Ragot, S.; Bossard, C.; Nadal, N.; et al. Ibrutinib, obinutuzumab, and venetoclax in relapsed and untreated patients with mantle cell lymphoma: A phase 1/2 trial. Blood 2021, 137, 877–887. [Google Scholar] [CrossRef]
- Zelenetz, A.D.; Salles, G.; Mason, K.D.; Casulo, C.; Le Gouill, S.; Sehn, L.H.; Tilly, H.; Cartron, G.; Chamuleau, M.E.D.; Goy, A.; et al. Venetoclax plus R- or G-CHOP in non-Hodgkin lymphoma: Results from the CAVALLI phase 1b trial. Blood 2019, 133, 1964–1976. [Google Scholar] [CrossRef]
- Morschhauser, F.; Feugier, P.; Flinn, I.W.; Gasiorowski, R.; Greil, R.; Illes, A.; Johnson, N.A.; Larouche, J.F.; Lugtenburg, P.J.; Patti, C.; et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. Blood 2021, 137, 600–609. [Google Scholar] [CrossRef] [PubMed]
- Zinzani, P.L.; Flinn, I.W.; Yuen, S.L.S.; Topp, M.S.; Rusconi, C.; Fleury, I.; Le Du, K.; Arthur, C.; Pro, B.; Gritti, G.; et al. Venetoclax-rituximab with or without bendamustine vs bendamustine-rituximab in relapsed/refractory follicular lymphoma. Blood 2020, 136, 2628–2637. [Google Scholar]
- Pullarkat, V.A.; Lacayo, N.J.; Jabbour, E.; Rubnitz, J.E.; Bajel, A.; Laetsch, T.W.; Leonard, J.; Colace, S.I.; Khaw, S.L.; Fleming, S.A.; et al. Venetoclax and Navitoclax in Combination with Chemotherapy in Patients with Relapsed or Refractory Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma. Cancer Discov. 2021, 11, 1440–1453. [Google Scholar] [CrossRef] [PubMed]
- Kadia, T.M.; Reville, P.K.; Borthakur, G.; Yilmaz, M.; Kornblau, S.; Alvarado, Y.; Dinardo, C.D.; Daver, N.; Jain, N.; Pemmaraju, N.; et al. Venetoclax plus intensive chemotherapy with cladribine, idarubicin, and cytarabine in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: A cohort from a single-centre, single-arm, phase 2 trial. Lancet Haematol. 2021, 8, e552–e561. [Google Scholar] [CrossRef]
- Garcia, J.S.; Kim, H.T.; Murdock, H.M.; Cutler, C.S.; Brock, J.; Gooptu, M.; Ho, V.T.; Koreth, J.; Nikiforow, S.; Romee, R.; et al. Adding venetoclax to fludarabine/busulfan RIC transplant for high-risk MDS and AML is feasible, safe, and active. Blood Adv. 2021, 5, 5536–5545. [Google Scholar] [CrossRef] [PubMed]
- King, A.L.O.; Mirza, F.N.; Lewis, J.M.; Carlson, K.R.; Huntington, S.; Foss, F.M.; Girardi, M. B-cell lymphoma 2 inhibitor venetoclax treatment of a patient with cutaneous T-cell lymphoma. JAAD Case Rep. 2021, 8, 89–92. [Google Scholar] [CrossRef] [PubMed]
- Cyrenne, B.M.; Lewis, J.M.; Weed, J.G.; Carlson, K.R.; Mirza, F.N.; Foss, F.M.; Girardi, M. Synergy of BCL2 and histone deacetylase inhibition against leukemic cells from cutaneous T-cell lymphoma patients. Blood 2017, 130, 2073–2083. [Google Scholar] [CrossRef]
- Xu, Y.; Ye, H. Progress in understanding the mechanisms of resistance to BCL-2 inhibitors. Exp. Hematol. Oncol. 2022, 11, 31. [Google Scholar] [CrossRef]
- Blombery, P.; Anderson, M.A.; Gong, J.N.; Thijssen, R.; Birkinshaw, R.W.; Thompson, E.R.; Teh, C.E.; Nguyen, T.; Xu, Z.; Flensburg, C.; et al. Acquisition of the Recurrent Gly101Val Mutation in BCL2 Confers Resistance to Venetoclax in Patients with Progressive Chronic Lymphocytic Leukemia. Cancer Discov. 2019, 9, 342–353. [Google Scholar] [CrossRef] [PubMed]
- Birkinshaw, R.W.; Gong, J.N.; Luo, C.S.; Lio, D.; White, C.A.; Anderson, M.A.; Blombery, P.; Lessene, G.; Majewski, I.J.; Thijssen, R.; et al. Structures of BCL-2 in complex with venetoclax reveal the molecular basis of resistance mutations. Nat. Commun. 2019, 10, 2385. [Google Scholar] [CrossRef]
- Tausch, E.; Close, W.; Dolnik, A.; Bloehdorn, J.; Chyla, B.; Bullinger, L.; Dohner, H.; Mertens, D.; Stilgenbauer, S. Venetoclax resistance and acquired BCL2 mutations in chronic lymphocytic leukemia. Haematologica 2019, 104, e434–e437. [Google Scholar] [CrossRef]
- Blombery, P.; Thompson, E.R.; Nguyen, T.; Birkinshaw, R.W.; Gong, J.N.; Chen, X.; McBean, M.; Thijssen, R.; Conway, T.; Anderson, M.A.; et al. Multiple BCL2 mutations cooccurring with Gly101Val emerge in chronic lymphocytic leukemia progression on venetoclax. Blood 2020, 135, 773–777. [Google Scholar] [CrossRef] [PubMed]
- Thangavadivel, S.; Byrd, J.C. Gly101Val BCL2 Mutation: One Step Closer to Understanding Venetoclax Resistance in CLL. Cancer Discov. 2019, 9, 320–322. [Google Scholar] [CrossRef] [PubMed]
- Lucas, F.; Larkin, K.; Gregory, C.T.; Orwick, S.; Doong, T.J.; Lozanski, A.; Lozanski, G.; Misra, S.; Ngankeu, A.; Ozer, H.G.; et al. Novel BCL2 mutations in venetoclax-resistant, ibrutinib-resistant CLL patients with BTK/PLCG2 mutations. Blood 2020, 135, 2192–2195. [Google Scholar] [CrossRef]
- Zhang, X.; Qian, J.; Wang, H.; Wang, Y.; Zhang, Y.; Qian, P.; Lou, Y.; Jin, J.; Zhu, H. Not BCL2 mutation but dominant mutation conversation contributed to acquired venetoclax resistance in acute myeloid leukemia. Biomark. Res. 2021, 9, 30. [Google Scholar] [CrossRef] [PubMed]
- Zhao, S.; Kanagal-Shamanna, R.; Navsaria, L.; Ok, C.Y.; Zhang, S.; Nomie, K.; Han, G.; Hao, D.; Hill, H.A.; Jiang, C.; et al. Efficacy of venetoclax in high risk relapsed mantle cell lymphoma (MCL)—Outcomes and mutation profile from venetoclax resistant MCL patients. Am. J. Hematol. 2020, 95, 623–629. [Google Scholar] [CrossRef]
- DiNardo, C.D.; Rausch, C.R.; Benton, C.; Kadia, T.; Jain, N.; Pemmaraju, N.; Daver, N.; Covert, W.; Marx, K.R.; Mace, M.; et al. Clinical experience with the BCL2-inhibitor venetoclax in combination therapy for relapsed and refractory acute myeloid leukemia and related myeloid malignancies. Am. J. Hematol. 2018, 93, 401–407. [Google Scholar] [CrossRef]
- Wang, Y.W.; Tsai, C.H.; Lin, C.C.; Tien, F.M.; Chen, Y.W.; Lin, H.Y.; Yao, M.; Lin, Y.C.; Lin, C.T.; Cheng, C.L.; et al. Cytogenetics and mutations could predict outcome in relapsed and refractory acute myeloid leukemia patients receiving BCL-2 inhibitor venetoclax. Ann. Hematol. 2020, 99, 501–511. [Google Scholar] [CrossRef]
- Cherry, E.M.; Abbott, D.; Amaya, M.; McMahon, C.; Schwartz, M.; Rosser, J.; Sato, A.; Schowinsky, J.; Inguva, A.; Minhajuddin, M.; et al. Venetoclax and azacitidine compared with induction chemotherapy for newly diagnosed patients with acute myeloid leukemia. Blood Adv. 2021, 5, 5565–5573. [Google Scholar] [CrossRef]
- Chow, S.; Tang, K.; Al-Abri, M.; Hall, V.; Tremblay-Lemay, R.; Rashedi, I.; Tsui, H.; Chan, S.M. RUNX1 mutations correlate with response to venetoclax combination therapies in relapsed/refractory acute myeloid leukemia. Leuk. Res. 2021, 111, 106735. [Google Scholar] [CrossRef]
- Wang, J.; Ye, X.; Fan, C.; Zhou, J.; Luo, S.; Jin, J.; Chen, D.; Zheng, Y.; Wu, C.; Zhu, X.; et al. Leukemia cutis with IDH1, DNMT3A and NRAS mutations conferring resistance to venetoclax plus 5-azacytidine in refractory AML. Biomark. Res. 2020, 8, 65. [Google Scholar] [CrossRef] [PubMed]
- Mill, C.P.; Fiskus, W.; DiNardo, C.D.; Birdwell, C.; Davis, J.A.; Kadia, T.M.; Takahashi, K.; Short, N.; Daver, N.; Ohanian, M.; et al. Effective therapy for AML with RUNX1 mutation by cotreatment with inhibitors of protein translation and BCL2. Blood 2022, 139, 907–921. [Google Scholar] [CrossRef]
- DiNardo, C.D.; Tiong, I.S.; Quaglieri, A.; MacRaild, S.; Loghavi, S.; Brown, F.C.; Thijssen, R.; Pomilio, G.; Ivey, A.; Salmon, J.M.; et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood 2020, 135, 791–803. [Google Scholar] [CrossRef]
- Stahl, M.; Menghrajani, K.; Derkach, A.; Chan, A.; Xiao, W.; Glass, J.; King, A.C.; Daniyan, A.F.; Famulare, C.; Cuello, B.M.; et al. Clinical and molecular predictors of response and survival following venetoclax therapy in relapsed/refractory AML. Blood Adv. 2021, 5, 1552–1564. [Google Scholar] [CrossRef]
- Fleischmann, M.; Scholl, S.; Frietsch, J.J.; Hilgendorf, I.; Schrenk, K.; Hammersen, J.; Prims, F.; Thiede, C.; Hochhaus, A.; Schnetzke, U. Clinical experience with venetoclax in patients with newly diagnosed, relapsed, or refractory acute myeloid leukemia. J. Cancer Res. Clin. Oncol. 2022, 148, 3191–3202. [Google Scholar] [CrossRef]
- Bisaillon, R.; Moison, C.; Thiollier, C.; Krosl, J.; Bordeleau, M.E.; Lehnertz, B.; Lavallee, V.P.; MacRae, T.; Mayotte, N.; Labelle, C.; et al. Genetic characterization of ABT-199 sensitivity in human AML. Leukemia 2020, 34, 63–74. [Google Scholar] [CrossRef] [PubMed]
- Tiong, I.S.; Dillon, R.; Ivey, A.; Teh, T.C.; Nguyen, P.; Cummings, N.; Taussig, D.C.; Latif, A.L.; Potter, N.E.; Runglall, M.; et al. Venetoclax induces rapid elimination of NPM1 mutant measurable residual disease in combination with low-intensity chemotherapy in acute myeloid leukaemia. Br. J. Haematol. 2021, 192, 1026–1030. [Google Scholar] [CrossRef] [PubMed]
- Lachowiez, C.A.; Loghavi, S.; Kadia, T.M.; Daver, N.; Borthakur, G.; Pemmaraju, N.; Naqvi, K.; Alvarado, Y.; Yilmaz, M.; Short, N.; et al. Outcomes of older patients with NPM1-mutated AML: Current treatments and the promise of venetoclax-based regimens. Blood Adv. 2020, 4, 1311–1320. [Google Scholar] [CrossRef] [PubMed]
- Zhu, H.H.; Qian, J.J.; Sun, W.J.; You, L.S.; Wang, Q.Q.; Naranmandura, H.; Jin, J. Venetoclax and arsenic showed synergistic anti-leukemia activity in vitro and in vivo for acute myeloid leukemia with the NPM1 mutation. Am. J. Hematol. 2020, 95, E55–E57. [Google Scholar] [CrossRef]
- Chan, S.M.; Thomas, D.; Corces-Zimmerman, M.R.; Xavy, S.; Rastogi, S.; Hong, W.J.; Zhao, F.; Medeiros, B.C.; Tyvoll, D.A.; Majeti, R. Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nat. Med. 2015, 21, 178–184. [Google Scholar] [CrossRef]
- Pollyea, D.A.; DiNardo, C.D.; Arellano, M.L.; Pigneux, A.; Fiedler, W.; Konopleva, M.; Rizzieri, D.A.; Smith, B.D.; Shinagawa, A.; Lemoli, R.M.; et al. Impact of Venetoclax and Azacitidine in Treatment-Naive Patients with Acute Myeloid Leukemia and IDH1/2 Mutations. Clin. Cancer Res. 2022, 28, 2753–2761. [Google Scholar] [CrossRef]
- Buege, M.J.; DiPippo, A.J.; DiNardo, C.D. Evolving Treatment Strategies for Elderly Leukemia Patients with IDH Mutations. Cancers 2018, 10, 187. [Google Scholar] [CrossRef] [PubMed]
- Jasra, S.; Kazemi, M.; Shah, N.; Chen, J.; Fehn, K.; Wang, Y.; Mantzaris, I.; Kornblum, N.; Sica, A.; Bachier, L.; et al. Case report of combination therapy with Azacytidine, Enasidenib and Venetoclax in primary refractory AML. Exp. Hematol. Oncol. 2021, 10, 1. [Google Scholar] [CrossRef]
- Cathelin, S.; Sharon, D.; Subedi, A.; Cojocari, D.; Phillips, D.C.; Leverson, J.D.; MacBeth, K.J.; Nicolay, B.; Narayanaswamy, R.; Ronseaux, S.; et al. Enasidenib-induced differentiation promotes sensitivity to venetoclax in IDH2-mutated acute myeloid leukemia. Leukemia 2022, 36, 869–872. [Google Scholar] [CrossRef]
- Venugopal, S.; Maiti, A.; DiNardo, C.D.; Loghavi, S.; Daver, N.G.; Kadia, T.M.; Rausch, C.R.; Alvarado, Y.; Ohanian, M.; Sasaki, K.; et al. Decitabine and venetoclax for IDH1/2-mutated acute myeloid leukemia. Am. J. Hematol. 2021, 96, E154–E157. [Google Scholar] [CrossRef]
- Venugopal, S.; Takahashi, K.; Daver, N.; Maiti, A.; Borthakur, G.; Loghavi, S.; Short, N.J.; Ohanian, M.; Masarova, L.; Issa, G.; et al. Efficacy and safety of enasidenib and azacitidine combination in patients with IDH2 mutated acute myeloid leukemia and not eligible for intensive chemotherapy. Blood Cancer J. 2022, 12, 10. [Google Scholar] [CrossRef] [PubMed]
- Aldoss, I.; Yang, D.; Pillai, R.; Sanchez, J.F.; Mei, M.; Aribi, A.; Ali, H.; Sandhu, K.; Al Malki, M.M.; Salhotra, A.; et al. Association of leukemia genetics with response to venetoclax and hypomethylating agents in relapsed/refractory acute myeloid leukemia. Am. J. Hematol. 2019, 94, E253–E255. [Google Scholar] [CrossRef]
- Nechiporuk, T.; Kurtz, S.E.; Nikolova, O.; Liu, T.; Jones, C.L.; D’Alessandro, A.; Culp-Hill, R.; d’Almeida, A.; Joshi, S.K.; Rosenberg, M.; et al. The TP53 Apoptotic Network Is a Primary Mediator of Resistance to BCL2 Inhibition in AML Cells. Cancer Discov. 2019, 9, 910–925. [Google Scholar] [CrossRef] [PubMed]
- Roberts, A.W.; Ma, S.; Kipps, T.J.; Coutre, S.E.; Davids, M.S.; Eichhorst, B.; Hallek, M.; Byrd, J.C.; Humphrey, K.; Zhou, L.; et al. Efficacy of venetoclax in relapsed chronic lymphocytic leukemia is influenced by disease and response variables. Blood 2019, 134, 111–122. [Google Scholar] [CrossRef] [PubMed]
- Tausch, E.; Schneider, C.; Robrecht, S.; Zhang, C.; Dolnik, A.; Bloehdorn, J.; Bahlo, J.; Al-Sawaf, O.; Ritgen, M.; Fink, A.M.; et al. Prognostic and predictive impact of genetic markers in patients with CLL treated with obinutuzumab and venetoclax. Blood 2020, 135, 2402–2412. [Google Scholar] [CrossRef]
- Kim, K.; Maiti, A.; Loghavi, S.; Pourebrahim, R.; Kadia, T.M.; Rausch, C.R.; Furudate, K.; Daver, N.G.; Alvarado, Y.; Ohanian, M.; et al. Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax. Cancer 2021, 127, 3772–3781. [Google Scholar] [CrossRef]
- Venugopal, S.; Shoukier, M.; Konopleva, M.; Dinardo, C.D.; Ravandi, F.; Short, N.J.; Andreeff, M.; Borthakur, G.; Daver, N.; Pemmaraju, N.; et al. Outcomes in patients with newly diagnosed TP53-mutated acute myeloid leukemia with or without venetoclax-based therapy. Cancer 2021, 127, 3541–3551. [Google Scholar] [CrossRef] [PubMed]
- Aldoss, I.; Zhang, J.; Pillai, R.; Shouse, G.; Sanchez, J.F.; Mei, M.; Nakamura, R.; Stein, A.S.; Forman, S.J.; Marcucci, G.; et al. Venetoclax and hypomethylating agents in TP53-mutated acute myeloid leukaemia. Br. J. Haematol. 2019, 187, e45–e48. [Google Scholar] [CrossRef]
- Chyla, B.; Daver, N.; Doyle, K.; McKeegan, E.; Huang, X.; Ruvolo, V.; Wang, Z.; Chen, K.; Souers, A.; Leverson, J.; et al. Genetic Biomarkers of Sensitivity and Resistance to Venetoclax Monotherapy in Patients with Relapsed Acute Myeloid Leukemia. Am. J. Hematol. 2018, 93, E202–E205. [Google Scholar] [CrossRef]
- Konopleva, M.; Thirman, M.J.; Pratz, K.W.; Garcia, J.S.; Recher, C.; Pullarkat, V.; Kantarjian, H.M.; DiNardo, C.D.; Dail, M.; Duan, Y.; et al. Impact of F LT3 Mutation on Outcomes after Venetoclax and Azacitidine for Patients with Treatment-Naive Acute Myeloid Leukemia. Clin. Cancer Res. 2022, 28, 2744–2752. [Google Scholar] [CrossRef] [PubMed]
- Aldoss, I.; Zhang, J.; Mei, M.; Al Malki, M.M.; Arslan, S.; Ngo, D.; Aribi, A.; Ali, H.; Sandhu, K.; Salhotra, A.; et al. Venetoclax and hypomethylating agents in FLT3-mutated acute myeloid leukemia. Am. J. Hematol. 2020, 95, 1193–1199. [Google Scholar] [CrossRef]
- Brinton, L.T.; Zhang, P.; Williams, K.; Canfield, D.; Orwick, S.; Sher, S.; Wasmuth, R.; Beaver, L.; Cempre, C.; Skinner, J.; et al. Synergistic effect of BCL2 and FLT3 co-inhibition in acute myeloid leukemia. J. Hematol. Oncol. 2020, 13, 139. [Google Scholar] [CrossRef] [PubMed]
- Singh Mali, R.; Zhang, Q.; DeFilippis, R.A.; Cavazos, A.; Kuruvilla, V.M.; Raman, J.; Mody, V.; Choo, E.F.; Dail, M.; Shah, N.P.; et al. Venetoclax combines synergistically with FLT3 inhibition to effectively target leukemic cells in FLT3-ITD+ acute myeloid leukemia models. Haematologica 2021, 106, 1034–1046. [Google Scholar] [CrossRef]
- Zhang, L.S.; Wang, J.; Xu, M.Z.; Wu, T.M.; Huang, S.M.; Cao, H.Y.; Sun, A.N.; Liu, S.B.; Xue, S.L. Rapid and Efficient Response to Gilteritinib and Venetoclax-Based Therapy in Two AML Patients with FLT3-ITD Mutation Unresponsive to Venetoclax Plus Azacitidine. OncoTargets Ther. 2022, 15, 159–164. [Google Scholar] [CrossRef]
- Post, S.M.; Ma, H.; Malaney, P.; Zhang, X.; Aitken, M.J.L.; Mak, P.Y.; Ruvolo, V.R.; Yasuhiro, T.; Kozaki, R.; Chan, L.E.; et al. AXL/MERTK inhibitor ONO-7475 potently synergizes with venetoclax and overcomes venetoclax resistance to kill FLT3-ITD acute myeloid leukemia. Haematologica 2021, 107, 1311–1322. [Google Scholar] [CrossRef] [PubMed]
- Gangat, N.; Guglielmelli, P.; Szuber, N.; Begna, K.H.; Patnaik, M.M.; Litzow, M.R.; Al-Kali, A.; Foran, J.M.; Palmer, J.M.; Alkhateeb, H.; et al. Venetoclax with azacitidine or decitabine in blast-phase myeloproliferative neoplasm: A multicenter series of 32 consecutive cases. Am. J. Hematol. 2021, 96, 781–789. [Google Scholar] [CrossRef] [PubMed]
- Morsia, E.; McCullough, K.; Joshi, M.; Cook, J.; Alkhateeb, H.B.; Al-Kali, A.; Begna, K.; Elliott, M.; Hogan, W.; Litzow, M.; et al. Venetoclax and hypomethylating agents in acute myeloid leukemia: Mayo Clinic series on 86 patients. Am. J. Hematol. 2020, 95, 1511–1521. [Google Scholar] [CrossRef]
- Pan, W.; Zhao, X.; Shi, W.; Jiang, Z.; Xiao, H. Venetoclax induced complete remission in extramedullary relapse of AML co-harboring NPM1, TET2, and NRAS mutations after haploidentical hematopoietic stem cell transplantation. Leuk. Lymphoma 2020, 61, 2756–2759. [Google Scholar] [CrossRef]
- Vaillant, F.; Merino, D.; Lee, L.; Breslin, K.; Pal, B.; Ritchie, M.E.; Smyth, G.K.; Christie, M.; Phillipson, L.J.; Burns, C.J.; et al. Targeting BCL-2 with the BH3 mimetic ABT-199 in estrogen receptor-positive breast cancer. Cancer Cell 2013, 24, 120–129. [Google Scholar] [CrossRef]
- Merino, D.; Lok, S.W.; Visvader, J.E.; Lindeman, G.J. Targeting BCL-2 to enhance vulnerability to therapy in estrogen receptor-positive breast cancer. Oncogene 2016, 35, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
- Muttiah, C.; Travers, A.; Whittle, J.R.; Dawson, S.-J.; Yeo, B.; Visvader, J.E.; Oakman, C.; Lindeman, G.J. Abstract OT-27-01: PALVEN: A phase 1b study of palbociclib, letrozole and venetoclax in estrogen receptor, BCL2-positive metastatic breast cancer. Cancer Res. 2021, 81, OT-27-01. [Google Scholar] [CrossRef]
- Lindeman, G.J.; Bowen, R.; Jerzak, K.J.; Song, X.; Decker, T.; Boyle, F.M.; McCune, S.L.; Armstrong, A.; Shannon, C.M.; Bertelli, G.; et al. Results from VERONICA: A randomized, phase II study of second-/third-line venetoclax (VEN) + fulvestrant (F) versus F alone in estrogen receptor (ER)-positive, HER2-negative, locally advanced, or metastatic breast cancer (LA/MBC). J. Clin. Oncol. 2021, 39, 1004. [Google Scholar] [CrossRef]
- Whittle, J.R.; Vaillant, F.; Surgenor, E.; Policheni, A.N.; Giner, G.; Capaldo, B.D.; Chen, H.R.; Liu, H.K.; Dekkers, J.F.; Sachs, N.; et al. Dual Targeting of CDK4/6 and BCL2 Pathways Augments Tumor Response in Estrogen Receptor-Positive Breast Cancer. Clin. Cancer Res. 2020, 26, 4120–4134. [Google Scholar] [CrossRef] [PubMed]
- Sun, Q.; Wang, Y.; Desgrosellier, J.S. Combined Bcl-2/Src inhibition synergize to deplete stem-like breast cancer cells. Cancer Lett. 2019, 457, 40–46. [Google Scholar] [CrossRef] [PubMed]
- Schroeder, B.; Vander Steen, T.; Espinoza, I.; Venkatapoorna, C.M.K.; Hu, Z.; Silva, F.M.; Regan, K.; Cuyas, E.; Meng, X.W.; Verdura, S.; et al. Fatty acid synthase (FASN) regulates the mitochondrial priming of cancer cells. Cell Death Dis. 2021, 12, 977. [Google Scholar] [CrossRef] [PubMed]
- Haikala, H.M.; Anttila, J.M.; Marques, E.; Raatikainen, T.; Ilander, M.; Hakanen, H.; Ala-Hongisto, H.; Savelius, M.; Balboa, D.; Von Eyss, B.; et al. Pharmacological reactivation of MYC-dependent apoptosis induces susceptibility to anti-PD-1 immunotherapy. Nat. Commun. 2019, 10, 620. [Google Scholar] [CrossRef]
- Alhoshani, A.; Alatawi, F.O.; Al-Anazi, F.E.; Attafi, I.M.; Zeidan, A.; Agouni, A.; El Gamal, H.M.; Shamoon, L.S.; Khalaf, S.; Korashy, H.M. BCL-2 Inhibitor Venetoclax Induces Autophagy-Associated Cell Death, Cell Cycle Arrest, and Apoptosis in Human Breast Cancer Cells. OncoTargets Ther. 2020, 13, 13357–13370. [Google Scholar] [CrossRef] [PubMed]
- Seveno, C.; Loussouarn, D.; Brechet, S.; Campone, M.; Juin, P.; Barille-Nion, S. gamma-Secretase inhibition promotes cell death, Noxa upregulation, and sensitization to BH3 mimetic ABT-737 in human breast cancer cells. Breast Cancer Res. 2012, 14, R96. [Google Scholar] [CrossRef]
- Aka, Y.; Karakas, B.; Acikbas, U.; Basaga, H.; Gul, O.; Kutuk, O. Kinome-wide RNAi screening for mediators of ABT-199 resistance in breast cancer cells identifies Wee1 as a novel therapeutic target. Int. J. Biochem. Cell Biol. 2021, 137, 106028. [Google Scholar] [CrossRef]
- Booth, L.; Roberts, J.L.; Avogadri-Connors, F.; Cutler, R.E., Jr.; Lalani, A.S.; Poklepovic, A.; Dent, P. The irreversible ERBB1/2/4 inhibitor neratinib interacts with the BCL-2 inhibitor venetoclax to kill mammary cancer cells. Cancer Biol. Ther. 2018, 19, 239–247. [Google Scholar] [CrossRef]
- Liu, D.; Qin, X.; Sun, Z.; Hou, S.; Lv, Q. Low DEDD expression in breast cancer cells indicates higher sensitivity to the Bcl-2-specific inhibitor ABT-199. Biochem. Biophys. Res. Commun. 2020, 525, 549–556. [Google Scholar] [CrossRef]
- Crump, L.S.; Wyatt, G.L.; Rutherford, T.R.; Richer, J.K.; Porter, W.W.; Lyons, T.R. Hormonal Regulation of Semaphorin 7a in ER+ Breast Cancer Drives Therapeutic Resistance. Cancer Res. 2021, 81, 187–198. [Google Scholar] [CrossRef]
- Young, A.; Bu, W.; Jiang, W.; Ku, A.; Kapali, J.; Dhamne, S.; Qin, L.; Hilsenbeck, S.G.; Du, Y.N.; Li, Y. Targeting the Pro-survival Protein BCL-2 to Prevent Breast Cancer. Cancer Prev. Res. 2022, 15, 3–10. [Google Scholar] [CrossRef]
- Goodwin, C.M.; Rossanese, O.W.; Olejniczak, E.T.; Fesik, S.W. Myeloid cell leukemia-1 is an important apoptotic survival factor in triple-negative breast cancer. Cell Death Differ. 2015, 22, 2098–2106. [Google Scholar] [CrossRef]
- Lucantoni, F.; Lindner, A.U.; O’Donovan, N.; Dussmann, H.; Prehn, J.H.M. Systems modeling accurately predicts responses to genotoxic agents and their synergism with BCL-2 inhibitors in triple negative breast cancer cells. Cell Death Dis. 2018, 9, 42. [Google Scholar] [CrossRef]
- Lochmann, T.L.; Floros, K.V.; Naseri, M.; Powell, K.M.; Cook, W.; March, R.J.; Stein, G.T.; Greninger, P.; Maves, Y.K.; Saunders, L.R.; et al. Venetoclax Is Effective in Small-Cell Lung Cancers with High BCL-2 Expression. Clin. Cancer Res. 2018, 24, 360–369. [Google Scholar] [CrossRef] [PubMed]
- Lam, L.T.; Lin, X.; Faivre, E.J.; Yang, Z.; Huang, X.; Wilcox, D.M.; Bellin, R.J.; Jin, S.; Tahir, S.K.; Mitten, M.; et al. Vulnerability of Small-Cell Lung Cancer to Apoptosis Induced by the Combination of BET Bromodomain Proteins and BCL2 Inhibitors. Mol. Cancer Ther. 2017, 16, 1511–1520. [Google Scholar] [CrossRef] [PubMed]
- Inoue-Yamauchi, A.; Jeng, P.S.; Kim, K.; Chen, H.C.; Han, S.; Ganesan, Y.T.; Ishizawa, K.; Jebiwott, S.; Dong, Y.; Pietanza, M.C.; et al. Targeting the differential addiction to anti-apoptotic BCL-2 family for cancer therapy. Nat. Commun. 2017, 8, 16078. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.Z.; Shilo, K.; Amann, J.M.; Shulman, A.; Hojjat-Farsangi, M.; Mellstedt, H.; Schultz, J.; Croce, C.M.; Carbone, D.P. Predicting ROR1/BCL2 combination targeted therapy of small cell carcinoma of the lung. Cell Death Dis. 2021, 12, 577. [Google Scholar] [CrossRef]
- Munkhbaatar, E.; Dietzen, M.; Agrawal, D.; Anton, M.; Jesinghaus, M.; Boxberg, M.; Pfarr, N.; Bidola, P.; Uhrig, S.; Hockendorf, U.; et al. MCL-1 gains occur with high frequency in lung adenocarcinoma and can be targeted therapeutically. Nat. Commun. 2020, 11, 4527. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Wang, L.; Zhang, Q.; Yan, Y.; Jiang, H.; Hu, R.; Zhou, X.; Liu, X.; Feng, J.; Lin, N. The Ibr-7 derivative of ibrutinib exhibits enhanced cytotoxicity against non-small cell lung cancer cells via targeting of mTORC1/S6 signaling. Mol. Oncol. 2019, 13, 946–958. [Google Scholar] [CrossRef]
- Kim, M.J.; Chen, G.; Sica, G.L.; Deng, X. Epigenetic modulation of FBW7/Mcl-1 pathway for lung cancer therapy. Cancer Biol. Ther. 2021, 22, 55–65. [Google Scholar] [CrossRef]
- Liu, Z.; Shah, N.; Marshall, K.L.; Sprowls, S.A.; Saralkar, P.; Mohammad, A.; Blethen, K.E.; Arsiwala, T.A.; Fladeland, R.; Lockman, P.R.; et al. Overcoming the acquired resistance to gefitinib in lung cancer brain metastasis in vitro and in vivo. Arch. Toxicol. 2021, 95, 3575–3587. [Google Scholar] [CrossRef]
- Potter, D.S.; Du, R.; Bhola, P.; Bueno, R.; Letai, A. Dynamic BH3 profiling identifies active BH3 mimetic combinations in non-small cell lung cancer. Cell Death Dis. 2021, 12, 741. [Google Scholar] [CrossRef]
- Park, D.; Anisuzzaman, A.S.M.; Magis, A.T.; Chen, G.; Xie, M.; Zhang, G.; Behera, M.; Sica, G.L.; Ramalingam, S.S.; Owonikoko, T.K.; et al. Discovery of Small Molecule Bak Activator for Lung Cancer Therapy. Theranostics 2021, 11, 8500–8516. [Google Scholar] [CrossRef]
- Tan, Y.; Li, X.; Tian, Z.; Chen, S.; Zou, J.; Lian, G.; Chen, S.; Huang, K.; Chen, Y. TIMP1 down-regulation enhances gemcitabine sensitivity and reverses chemoresistance in pancreatic cancer. Biochem. Pharmacol. 2021, 189, 114085. [Google Scholar] [CrossRef]
- Zhou, Y.; Liu, H.; Xue, R.; Tang, W.; Zhang, S. BH3 Mimetic ABT-199 Enhances the Sensitivity of Gemcitabine in Pancreatic Cancer in vitro and in vivo. Dig. Dis. Sci. 2018, 63, 3367–3375. [Google Scholar] [CrossRef]
- Jakubowska, M.A.; Kerkhofs, M.; Martines, C.; Efremov, D.G.; Gerasimenko, J.V.; Gerasimenko, O.V.; Petersen, O.H.; Bultynck, G.; Vervliet, T.; Ferdek, P.E. ABT-199 (Venetoclax), a BH3-mimetic Bcl-2 inhibitor, does not cause Ca(2+)-signalling dysregulation or toxicity in pancreatic acinar cells. Br. J. Pharmacol. 2019, 176, 4402–4415. [Google Scholar] [CrossRef]
- King, H.M.; Rana, S.; Kubica, S.P.; Mallareddy, J.R.; Kizhake, S.; Ezell, E.L.; Zahid, M.; Naldrett, M.J.; Alvarez, S.; Law, H.C.; et al. Aminopyrazole based CDK9 PROTAC sensitizes pancreatic cancer cells to venetoclax. Bioorg. Med. Chem. Lett. 2021, 43, 128061. [Google Scholar] [CrossRef]
- Morimoto, Y.; Takada, K.; Takeuchi, O.; Watanabe, K.; Hirohara, M.; Hamamoto, T.; Masuda, Y. Bcl-2/Bcl-xL inhibitor navitoclax increases the antitumor effect of Chk1 inhibitor prexasertib by inducing apoptosis in pancreatic cancer cells via inhibition of Bcl-xL but not Bcl-2. Mol. Cell. Biochem. 2020, 472, 187–198. [Google Scholar] [CrossRef]
- Kour, S.; Rana, S.; Contreras, J.I.; King, H.M.; Robb, C.M.; Sonawane, Y.A.; Bendjennat, M.; Crawford, A.J.; Barger, C.J.; Kizhake, S.; et al. CDK5 Inhibitor Downregulates Mcl-1 and Sensitizes Pancreatic Cancer Cell Lines to Navitoclax. Mol. Pharmacol. 2019, 96, 419–429. [Google Scholar] [CrossRef] [PubMed]
- Lafontaine, J.; Cardin, G.B.; Malaquin, N.; Boisvert, J.S.; Rodier, F.; Wong, P. Senolytic Targeting of Bcl-2 Anti-Apoptotic Family Increases Cell Death in Irradiated Sarcoma Cells. Cancers 2021, 13, 386. [Google Scholar] [CrossRef] [PubMed]
- Muenchow, A.; Weller, S.; Hinterleitner, C.; Malenke, E.; Bugl, S.; Wirths, S.; Muller, M.R.; Schulze-Osthoff, K.; Aulitzky, W.E.; Kopp, H.G.; et al. The BCL-2 selective inhibitor ABT-199 sensitizes soft tissue sarcomas to proteasome inhibition by a concerted mechanism requiring BAX and NOXA. Cell Death Dis. 2020, 11, 701. [Google Scholar] [CrossRef]
- Heinicke, U.; Haydn, T.; Kehr, S.; Vogler, M.; Fulda, S. BCL-2 selective inhibitor ABT-199 primes rhabdomyosarcoma cells to histone deacetylase inhibitor-induced apoptosis. Oncogene 2018, 37, 5325–5339. [Google Scholar] [CrossRef] [PubMed]
- Fairchild, C.K., Jr.; Floros, K.V.; Jacob, S.; Coon, C.M.; Puchalapalli, M.; Hu, B.; Harada, H.; Dozmorov, M.G.; Koblinski, J.E.; Smith, S.C.; et al. Unmasking BCL-2 Addiction in Synovial Sarcoma by Overcoming Low NOXA. Cancers 2021, 13, 2310. [Google Scholar] [CrossRef] [PubMed]
- Heisey, D.A.R.; Lochmann, T.L.; Floros, K.V.; Coon, C.M.; Powell, K.M.; Jacob, S.; Calbert, M.L.; Ghotra, M.S.; Stein, G.T.; Maves, Y.K.; et al. The Ewing Family of Tumors Relies on BCL-2 and BCL-XL to Escape PARP Inhibitor Toxicity. Clin. Cancer Res. 2019, 25, 1664–1675. [Google Scholar] [CrossRef]
- Hoda, M.A.; Pirker, C.; Dong, Y.; Schelch, K.; Heffeter, P.; Kryeziu, K.; van Schoonhoven, S.; Klikovits, T.; Laszlo, V.; Rozsas, A.; et al. Trabectedin Is Active against Malignant Pleural Mesothelioma Cell and Xenograft Models and Synergizes with Chemotherapy and Bcl-2 Inhibition In Vitro. Mol. Cancer Ther. 2016, 15, 2357–2369. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, Y.; Fan, X.; Song, J.; Wu, H.; Han, J.; Lu, L.; Weng, X.; Nie, G. ABT-199-mediated inhibition of Bcl-2 as a potential therapeutic strategy for nasopharyngeal carcinoma. Biochem. Biophys. Res. Commun. 2018, 503, 1214–1220. [Google Scholar] [CrossRef]
- Yin, L.; Zeng, Y.; Zeng, R.; Chen, Y.; Wang, T.L.; Rodabaugh, K.J.; Yu, F.; Natarajan, A.; Karpf, A.R.; Dong, J. Protein kinase RNA-activated controls mitotic progression and determines paclitaxel chemosensitivity through B-cell lymphoma 2 in ovarian cancer. Oncogene 2021, 40, 6772–6785. [Google Scholar] [CrossRef]
- Perimenis, P.; Galaris, A.; Voulgari, A.; Prassa, M.; Pintzas, A. IAP antagonists Birinapant and AT-406 efficiently synergise with either TRAIL, BRAF, or BCL-2 inhibitors to sensitise BRAFV600E colorectal tumour cells to apoptosis. BMC Cancer 2016, 16, 624. [Google Scholar] [CrossRef] [PubMed]
- O’Farrell, A.C.; Jarzabek, M.A.; Lindner, A.U.; Carberry, S.; Conroy, E.; Miller, I.S.; Connor, K.; Shiels, L.; Zanella, E.R.; Lucantoni, F.; et al. Implementing Systems Modelling and Molecular Imaging to Predict the Efficacy of BCL-2 Inhibition in Colorectal Cancer Patient-Derived Xenograft Models. Cancers 2020, 12, 2978. [Google Scholar] [CrossRef] [PubMed]
- Yu, Z.; Du, J.; Zhao, Y.; Gao, Y.; Li, Y.; Zhao, K.; Lu, N. A novel kinase inhibitor, LZT-106, downregulates Mcl-1 and sensitizes colorectal cancer cells to BH3 mimetic ABT-199 by targeting CDK9 and GSK-3beta signaling. Cancer Lett. 2021, 498, 31–41. [Google Scholar] [CrossRef]
- Huang, Q.; He, S.; Zhan, D. Osimertinib is a dual inhibitor of hepatocellular carcinoma and angiogenesis in an EGFR-independent manner, and synergizes with venetoclax. J. Cancer Res. Clin. Oncol. 2023, 149, 10727–10735. [Google Scholar] [CrossRef] [PubMed]
- Yu, X.; Dobrikov, M.; Keir, S.T.; Gromeier, M.; Pastan, I.H.; Reisfeld, R.; Bigner, D.D.; Chandramohan, V. Synergistic antitumor effects of 9.2.27-PE38KDEL and ABT-737 in primary and metastatic brain tumors. PLoS ONE 2019, 14, e0210608. [Google Scholar] [CrossRef]
- Adamova, B.; Rihova, K.; Pokludova, J.; Benes, P.; Smarda, J.; Navratilova, J. Synergistic cytotoxicity of perifosine and ABT-737 to colon cancer cells. J. Cell. Mol. Med. 2023, 27, 76–88. [Google Scholar] [CrossRef] [PubMed]
- Zheng, R.; You, Z.; Jia, J.; Lin, S.; Han, S.; Liu, A.; Long, H.; Wang, S. Curcumin enhances the antitumor effect of ABT-737 via activation of the ROS-ASK1-JNK pathway in hepatocellular carcinoma cells. Mol. Med. Rep. 2016, 13, 1570–1576. [Google Scholar] [CrossRef] [PubMed]
- Ren, J.; Li, G.; Zhao, W.; Lin, L.; Ye, T. Norcantharidin combined with ABT-737 for hepatocellular carcinoma: Therapeutic effects and molecular mechanisms. World J. Gastroenterol. 2016, 22, 3962–3968. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Xu, J.; Dong, X.; Huang, D.C.S.; Xu, P.; Zhao, Q.; Chen, B. Current Advances and Future Strategies for BCL-2 Inhibitors: Potent Weapons against Cancers. Cancers 2023, 15, 4957. https://doi.org/10.3390/cancers15204957
Xu J, Dong X, Huang DCS, Xu P, Zhao Q, Chen B. Current Advances and Future Strategies for BCL-2 Inhibitors: Potent Weapons against Cancers. Cancers. 2023; 15(20):4957. https://doi.org/10.3390/cancers15204957
Chicago/Turabian StyleXu, Jiaxuan, Xiaoqing Dong, David C. S. Huang, Peipei Xu, Quan Zhao, and Bing Chen. 2023. "Current Advances and Future Strategies for BCL-2 Inhibitors: Potent Weapons against Cancers" Cancers 15, no. 20: 4957. https://doi.org/10.3390/cancers15204957
APA StyleXu, J., Dong, X., Huang, D. C. S., Xu, P., Zhao, Q., & Chen, B. (2023). Current Advances and Future Strategies for BCL-2 Inhibitors: Potent Weapons against Cancers. Cancers, 15(20), 4957. https://doi.org/10.3390/cancers15204957