Abstract 940: The combinatorial activity of Eftozanermin ABBV621, a novel and potent TRAIL receptor agonist fusion protein, in pre-clinical models of hematologic malignancies

Author(s):  
Morey Smith ◽  
Sha Jin ◽  
Dong Cheng ◽  
Haichao Zhang ◽  
Jason Huska ◽  
...  
Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 41-41
Author(s):  
Morey L Smith ◽  
Sha Jin ◽  
Dong Chen ◽  
Haichao Zhang ◽  
Jason Huska ◽  
...  

Cell death can be initiated through activation of the extrinsic and intrinsic apoptotic signaling pathways. Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily of cytokines, preferentially triggers the extrinsic apoptotic pathway by binding as a trimer to two closely related cell surface death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5). Receptor trimerization leads to the formation of the death-inducing signaling complex (DISC) to recruit and activate downstream caspases that ultimately leads to apoptotic cell death. Because TRAIL signaling induces apoptosis, several TRAIL receptor agonists have been developed for the treatment of cancer. ABBV-621 is a novel, second generation TRAIL receptor agonist that is an engineered fusion protein consisting of an IgG1-Fc linked to a single chain trimer of TRAIL subunits resulting in a total of six death receptor binding sites per molecule to maximize receptor clustering that is currently being tested in Phase I clinical trials (NCT03082209). To expand upon the potential therapeutic utility of ABBV-621, we tested the combinatorial activity of ABBV-621 with numerous standard-of-care (SoC) therapeutics and targeted agents in diffuse large B-cell lymphoma (DLBCL), acute myeloid leukemia (AML) and multiple myeloma (MM) cell lines. Thein vitroresults led to selection of agents to combine with ABBV-621 forin vivostudies. In DLBCL cell line-derived xenograft (CDX) preclinical models, we observed combination activity of ABBV-621 with pevonedistat (PEV) a selective NEDD8 inhibitor. Additionally, synergistic activity was observed with ABBV-621 with either bendamustine (BED) or rituximab (RTX) alone, or BED/RTX together. In AML, we observed compelling combination activity of ABBV-621 with PEV in cell line-derived xenograft (CDX) models. In MM, combination of ABBV-621 plus bortezomib (BTZ) resulted in deeper anti-tumorigenic activity than either agent alone in several CDX models. The pre-clinical data presented here support expanding the indications and settings where ABBV-621 may have utility. A clinical trial assessing the activity of ABBV-621 in combination with bortezomib and dexamethasone in R/R MM patients is planned. Disclosures: All authors are employees of AbbVie. The design, study conduct, and financial support for this research were provided by AbbVie. AbbVie participated in the interpretation of data, review, and approval of the publication. Disclosures Smith: AbbVie:Current Employment, Current equity holder in publicly-traded company.Jin:AbbVie:Current Employment, Current equity holder in publicly-traded company.Chen:AbbVie:Current Employment, Current equity holder in publicly-traded company.Zhang:AbbVie:Current Employment, Current equity holder in publicly-traded company.Huska:AbbVie:Current Employment, Current equity holder in publicly-traded company.Widomski:AbbVie:Current Employment, Current equity holder in publicly-traded company.Bontcheva:AbbVie:Current Employment, Current equity holder in publicly-traded company.Buchanan:AbbVie:Current Employment, Current equity holder in publicly-traded company.Morgan-Lappe:AbbVie:Current Employment, Current equity holder in publicly-traded company.Phillips:AbbVie:Current Employment, Current equity holder in publicly-traded company.Tahir:AbbVie:Current Employment, Current equity holder in publicly-traded company.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 3013-3013 ◽  
Author(s):  
Mark J. Ratain ◽  
Toshihiko Doi ◽  
Maja J. De Jonge ◽  
Patricia LoRusso ◽  
Martin Dunbar ◽  
...  

3013 Background: ABBV-621 is a potent tumor-necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor agonist fusion protein that induces apoptotic cell death, particularly in DR4/5 expressing tumor models. Methods: Patients (pts) with previously treated solid tumors and ECOG 0–2 were administered ABBV-621 (2.5–15 mg/kg IV) on day (D) 1 (dose level [DL] 1) or D1D8 (DL2 and beyond) of each 21-day cycle. Dose escalation (DE) was guided by a Bayesian continual reassessment method. In addition to PK studies, blood-based PD markers of apoptosis (M30, M65) and drug binding were assessed. Results: As of 14 December 2018, 57 pts were enrolled in the DE portion, of which 30% had pancreatic, 23% colorectal cancer, and 47% other tumor types; 13 were KRAS mutant. Median age was 61 yrs. 60% were male; pts had a median of 4 prior regimens (range 1–10). Pts per DL: 2.5 (5 on D1, 16 on D1D8), 3.75 (12), 5 (6), 6.5 (6), 8.5 (4), 11 (4), and 15 mg/kg (4). Median duration of ABBV-621 exposure was 2 cycles (range 1–11). Seven pts had dose-limiting toxicities: respiratory failure (5 mg/kg; Grade 5, the only treatment-related death), blood bilirubin increased (3.75, 6.5 mg/kg), nausea (3.75 mg/kg), fatigue (3.75 mg/kg), increased ALT (2.5, 3.75, 6.5, 15 mg/kg), and increased AST (6.5 mg/kg). Summary of AEs is shown in Table. Clinical trial information: NCT03082209. A partial response (duration 20 weeks) was observed in a pt with pancreatic cancer (2.5 mg/kg D1D8). 27 pts had stable disease (6 pts for > 12 weeks). ABBV-621 PK was linear (mean ± SD clearance was 1.79 mL/h/kg ± 0.44) with a terminal half-life of 36.7 ± 5.55 h (n = 49). ABBV-621 bound to decoy receptors on neutrophils for up to 168 h; the duration of binding was dose-dependent. M30 and M65 increased at 8, 24, and 48 h following ABBV-621, but effect was independent of dose. Conclusions: ABBV-621 shows evidence of antitumor activity and effect on blood-based markers of apoptosis, with acceptable toxicity (MTD not reached). NCT03082209.[Table: see text]


2011 ◽  
Author(s):  
Junaid Abdulghani ◽  
Joshua E. Allen ◽  
David T. Dicker ◽  
Charles D. Smith ◽  
Robin C. Humphreys ◽  
...  

2019 ◽  
Author(s):  
Andrew J. Sawyer ◽  
Sara Ghassemifar ◽  
Christina Wong ◽  
Jennifer Richards ◽  
Stephanie Grabow ◽  
...  

Oncogene ◽  
2008 ◽  
Vol 27 (2) ◽  
pp. 208-217 ◽  
Author(s):  
D E Spaner ◽  
R Foley ◽  
J Galipeau ◽  
J Bramson

2019 ◽  
Author(s):  
Andrew J. Sawyer ◽  
Sara Ghassemifar ◽  
Christina Wong ◽  
Jennifer Richards ◽  
Stephanie Grabow ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1475-1475
Author(s):  
Myron S. Czuczman ◽  
Sreeram Maddipatla ◽  
Joy Knight ◽  
Francisco J. Hernandez-Ilizaliturri

Abstract Tumor necrosis factor-related apoptosis ligand (TRAIL) or agonist antibodies targeting specific TRAIL-receptors possess significant in vitro and in vivo anti-tumor activity. HGS-ETR1 (mapatumumab) and HGS-ETR2 are two fully human recombinant, high affinity IgG1λ mAbs targeting TRAIL-R1 (Death-receptor 4, DR-4) and TRAIL-R2 (Death-receptor 5, DR-5), respectively. TRAIL-receptor agonist mAbs trigger the extrinsic (mitochondria-independent) apoptotic pathway and enhance the anti-tumor effects of chemotherapy drugs against NHL. Combining TRAIL-receptor agonist mAbs with biological agents (e.g. rituximab) capable of activating the intrinsic (mitochondria-dependent) apoptotic pathway may result in synergistic effects against NHL. Objective: To study the biological effects of rituximab when combined with either HGS-ETR1 or HGS-ETR2. Materials and methods: A panel of NHL cell lines [Raji, SUDHL-4, SU-DHL-10, Ramos, and two rituximab resistant cell lines (RRCL), 2R and 4RH] were utilized. Expression of CD20, TRAIL-R1 and TRAIL-R2 in the NHL cells used were evaluated by flow cytometry. NHL cells were exposed to HGS-ETR1 or HGS-ETR2 followed by either rituximab, istoype control antibody or media alone. DNA synthesis and cell growth arrest were quantified by [H3] Thymidine incorporation assays at 24 and 48 hrs. Induction of apoptosis was detected by multiparameter flow cytometric analysis. For antibody-dependent cellular cytotoxicity (ADCC)/Complement mediated cytotoxicity (CMC) studies, 51Cr-labeled NHL cells were exposed to HGS-ETR1 or HGS-ETR2 (5μg/ml) +/− rituximab or isotype control antibody (10μg/ml) and peripheral blood mononuclear cells (Effector: Target ratio 40:1) or human serum, respectively. 51Cr-release was measured and the percentage of lysis was calculated. Statistical analysis of results was performed using the Chi-square test. Results: In sensitive cells in vitro exposure to HGS-ETR1 resulted in significant apoptosis (30–50%) and decrease in DNA synthesis. The combination of HGS-ETR1 and rituximab resulted in significant inhibition of cell proliferation (90% reduction) when compared to either HGS-ETR1 (60% reduction) or rituximab (5% reduction) alone at 24 and 48 hrs. In sensitive cells HGS-ETR1 induced CMC and ADCC as a single agent. No significant anti-tumor activity was observed with HGS-ETR2. The biological activity of anti-TRAIL receptors mAbs did not correlate to TRAIL-R1 or TRAIL-R2 cell surface antigen expression. Conclusions: Targeting TRAIL-R1 with HGS-ETR1 induces apoptosis, cell growth arrest and in some cell lines ADCC/CMC. The combination of HGS-ETR1 with rituximab results in significant synergistic activity (i.e. anti-proliferation) and warrants further pre-clinical and clinical evaluation in B-cell lymphomas.


PLoS ONE ◽  
2013 ◽  
Vol 8 (9) ◽  
pp. e75414 ◽  
Author(s):  
Junaid Abdulghani ◽  
Joshua E. Allen ◽  
David T. Dicker ◽  
Yingqiu Yvette Liu ◽  
David Goldenberg ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document