Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Edward B. Reilly is active.

Publication


Featured researches published by Edward B. Reilly.


Molecular Cancer Therapeutics | 2016

ABT-414, an Antibody Drug Conjugate Targeting a Tumor-Selective EGFR Epitope

Andrew C. Phillips; Erwin R. Boghaert; Kedar S. Vaidya; Michael J. Mitten; Suzanne M. Norvell; Hugh D. Falls; Peter J. DeVries; Dong Cheng; Jonathan A. Meulbroek; Fritz G. Buchanan; Laura M. McKay; Neal Goodwin; Edward B. Reilly

Targeting tumor-overexpressed EGFR with an antibody–drug conjugate (ADC) is an attractive therapeutic strategy; however, normal tissue expression represents a significant toxicity risk. The anti-EGFR antibody ABT-806 targets a unique tumor-specific epitope and exhibits minimal reactivity to EGFR in normal tissue, suggesting its suitability for the development of an ADC. We describe the binding properties and preclinical activity of ABT-414, an ABT-806 monomethyl auristatin F conjugate. In vitro, ABT-414 selectively kills tumor cells overexpressing wild-type or mutant forms of EGFR. ABT-414 inhibits the growth of xenograft tumors with high EGFR expression and causes complete regressions and cures in the most sensitive models. Tumor growth inhibition is also observed in tumor models with EGFR mutations, including activating mutations and those with the exon 2–7 deletion [EGFR variant III (EGFRvIII)], commonly found in glioblastoma multiforme. ABT-414 exhibits potent cytotoxicity against glioblastoma multiforme patient-derived xenograft models expressing either wild-type EGFR or EGFRvIII, with sustained regressions and cures observed at clinically relevant doses. ABT-414 also combines with standard-of-care treatment of radiation and temozolomide, providing significant therapeutic benefit in a glioblastoma multiforme xenograft model. On the basis of these results, ABT-414 has advanced to phase I/II clinical trials, and objective responses have been observed in patients with both amplified wild-type and EGFRvIII-expressing tumors. Mol Cancer Ther; 15(4); 661–9. ©2016 AACR.


Journal of Immunology | 2008

The Potency of Erythropoietin-Mimic Antibodies Correlates Inversely with Affinity

Susan E. Lacy; Peter J. DeVries; Nancy Xie; Emma Fung; Richard R. Lesniewski; Edward B. Reilly

Preclinical animal studies have shown that Ab12.6, an agonistic human Ab targeting the erythropoietin receptor (EPOR), exhibits several potential dosing and safety features that make it an attractive clinical candidate for the treatment of anemia. Ab12.6 was derived by yeast display affinity maturation of parental Ab12, a strategy initially intended to improve off-rate and affinity for EPOR, thereby enhancing erythropoietic activity. Analysis of full-length IgGs derived from yeast clones identified sequences within Ab12 CDRH2 that independently influenced both affinity and potency. The Ab12.6 derivative displayed improved in vitro potency and in vivo efficacy, although its binding affinity to the EPOR was lower than that of the parent Ab12. Additional Ab12 derivatives also exhibited an inverse correlation between affinity and potency. These results suggest that for this class of agonistic Abs, faster off-rates may permit continuous receptor stimulation and more efficient erythropoiesis.


Molecular Cancer Therapeutics | 2015

Characterization of ABT-806, a Humanized Tumor-Specific Anti-EGFR Monoclonal Antibody

Edward B. Reilly; Andrew C. Phillips; Fritz G. Buchanan; Gillian A. Kingsbury; Yumin Zhang; Jonathan A. Meulbroek; Todd B. Cole; Peter J. DeVries; Hugh D. Falls; Christine Beam; Jinming Gu; Enrico L. Digiammarino; Joann P. Palma; Cherrie K. Donawho; Neal C. Goodwin; Andrew M. Scott

Despite clinical efficacy, current approved agents targeting EGFR are associated with on-target toxicities as a consequence of disrupting normal EGFR function. MAb 806 is a novel EGFR antibody that selectively targets a tumor-selective epitope suggesting that a mAb 806-based therapeutic would retain antitumor activity without the on-target toxicities associated with EGFR inhibition. To enable clinical development, a humanized variant of mAb 806 designated ABT-806 was generated and is currently in phase 1 trials. We describe the characterization of binding and functional properties of ABT-806 compared with the clinically validated anti-EGFR antibody cetuximab. ABT-806 binds the mutant EGFRvIII with high affinity and, relative to cetuximab, exhibits increased potency against glioblastoma multiforme cell line and patient-derived xenografts expressing this form of the receptor. ABT-806 also inhibits the growth of squamous cell carcinoma xenograft models expressing high levels of wild-type EGFR, associated with inhibition of EGFR signaling, although higher doses of ABT-806 than cetuximab are required for similar activity. ABT-806 enhances in vivo potency of standard-of-care therapies used to treat glioblastoma multiforme and head and neck squamous cell carcinoma. An indium-labeled version of ABT-806, [111In]-ABT-806, used to investigate the relationship between dose and receptor occupancy, revealed greater receptor occupancy at lowers doses in an EGFRvIII-expressing model and significant uptake in an orthotopic model. Collectively, these results suggest that ABT-806 may have antitumor activity superior to cetuximab in EGFRvIII-expressing tumors, and similar activity to cetuximab in tumors highly overexpressing wild-type EGFR with reduced toxicity. Mol Cancer Ther; 14(5); 1141–51. ©2015 AACR.


Bioorganic & Medicinal Chemistry Letters | 2001

Discovery and SAR of diarylsulfide cyclopropylamide LFA-1/ICAM-1 interaction antagonists

James T. Link; Bryan K. Sorensen; Gang Liu; Zhonghua Pei; Edward B. Reilly; Sandra Leitza; Greg Okasinski

Diarylsulfide cyclopropylamides were synthesized and evaluated as LFA-1/ICAM-1 interaction antagonists. A substituent pattern was identified which maximized potency and minimized protein binding as exemplified by antagonist 30 (IC50 = 5 nM).


BMC Cancer | 2016

Anti-c-Met monoclonal antibody ABT-700 breaks oncogene addiction in tumors with MET amplification.

Jieyi Wang; Liliane Goetsch; Lora A. Tucker; Qian Zhang; Alexandra Gonzalez; Kedar S. Vaidya; Anatol Oleksijew; Erwin R. Boghaert; Minghao Song; Irina A. Sokolova; Ekaterina Pestova; Mark Gerald Anderson; William N. Pappano; Peter Ansell; Anahita Bhathena; Louie Naumovski; Nathalie Corvaia; Edward B. Reilly

Backgroundc-Met is the receptor tyrosine kinase for hepatocyte growth factor (HGF) encoded by the MET proto-oncogene. Aberrant activation of c-Met resulting from MET amplification and c-Met overexpression is associated with poor clinical outcome in multiple malignancies underscoring the importance of c-Met signaling in cancer progression. Several c-Met inhibitors have advanced to the clinic; however, the development of inhibitory c-Met-directed therapeutic antibodies has been hampered by inherent agonistic activity.MethodWe generated and tested a bivalent anti-c-Met monoclonal antibody ABT-700 in vitro for binding potency and antagonistic activity and in vivo for antitumor efficacy in human tumor xenografts. Human cancer cell lines and gastric cancer tissue microarrays were examined for MET amplification by fluorescence in situ hybridization (FISH).ResultsABT-700 exhibits a distinctive ability to block both HGF-independent constitutive c-Met signaling and HGF-dependent activation of c-Met. Cancer cells addicted to the constitutively activated c-Met signaling driven by MET amplification undergo apoptosis upon exposure to ABT-700. ABT-700 induces tumor regression and tumor growth delay in preclinical tumor models of gastric and lung cancers harboring amplified MET. ABT-700 in combination with chemotherapeutics also shows additive antitumor effect. Amplification of MET in human cancer tissues can be identified by FISH.ConclusionsThe preclinical attributes of ABT-700 in blocking c-Met signaling, inducing apoptosis and suppressing tumor growth in cancers with amplified MET provide rationale for examining its potential clinical utility for the treatment of cancers harboring MET amplification.


Clinical Cancer Research | 2017

ABBV-399, a c-Met Antibody Drug Conjugate that Targets Both MET Amplified and c-Met Overexpressing Tumors, Irrespective of MET Pathway Dependence.

Jieyi Wang; Mark G. Anderson; Anatol Oleksijew; Kedar S. Vaidya; Erwin R. Boghaert; Lora A. Tucker; Qian Zhang; Edward K. Han; Joann P. Palma; Louie Naumovski; Edward B. Reilly

Purpose: Despite the importance of the MET oncogene in many malignancies, clinical strategies targeting c-Met have benefitted only small subsets of patients with tumors driven by signaling through the c-Met pathway, thereby necessitating selection of patients with MET amplification and/or c-Met activation most likely to respond. An ADC targeting c-Met could overcome these limitations with potential as a broad-acting therapeutic. Experimental Design: ADC ABBV-399 was generated with the c-Met–targeting antibody, ABT-700. Antitumor activity was evaluated in cancer cells with overexpressed c-Met or amplified MET and in xenografts including patient-derived xenograft (PDX) models and those refractory to other c-Met inhibitors. The correlation between c-Met expression and sensitivity to ABBV-399 in tumor and normal cell lines was assessed to evaluate the risk of on-target toxicity. Results: A threshold level of c-Met expressed by sensitive tumor but not normal cells is required for significant ABBV-399–mediated killing of tumor cells. Activity extends to c-Met or amplified MET cell line and PDX models where significant tumor growth inhibition and regressions are observed. ABBV-399 inhibits growth of xenograft tumors refractory to other c-Met inhibitors and provides significant therapeutic benefit in combination with standard-of-care chemotherapy. Conclusions: ABBV-399 represents a novel therapeutic strategy to deliver a potent cytotoxin to c-Met–overexpressing tumor cells enabling cell killing regardless of reliance on MET signaling. ABBV-399 has progressed to a phase I study where it has been well tolerated and has produced objective responses in c-Met–expressing non–small cell lung cancer (NSCLC) patients. Clin Cancer Res; 23(4); 992–1000. ©2016 AACR.


Cancer | 2018

Efficacy and safety results of depatuxizumab mafodotin (ABT-414) in patients with advanced solid tumors likely to overexpress epidermal growth factor receptor: Depatux-M in EGFR-Overexpressing Tumors

Glenwood D. Goss; Everett E. Vokes; Michael S. Gordon; Leena Gandhi; Kyriakos P. Papadopoulos; Drew W. Rasco; JuDee Fischer; Katharine L. Chu; William Ames; Rajendar K. Mittapalli; Ho-Jin Lee; Jiewei Zeng; Lisa Roberts-Rapp; Lise I. Loberg; Peter Ansell; Edward B. Reilly; Christopher Ocampo; Kyle D. Holen; Anthony W. Tolcher

Epidermal growth factor receptor (EGFR) alterations are associated with multiple cancers. Current EGFR‐directed therapies have led to increased efficacy but are associated with specific side effects. The antibody‐drug conjugate depatuxizumab mafodotin (depatux‐m) targets EGFR with a monoclonal antibody linked to a cytotoxin, and is highly tumor‐specific.


Molecular Cancer Therapeutics | 2013

Abstract A250: ABT-414: An anti-EGFR antibody-drug conjugate as a potential therapeutic for the treatment of patients with squamous cell tumors.

Andrew C. Phillips; Erwin R. Boghaert; Kedar S. Vaidya; Peter Ansell; David R. Shalinsky; Yumin Zhang; Martin J. Voorbach; Sarah R. Mudd; Kyle D. Holen; Rod Humerickhouse; Edward B. Reilly

ABT-414 is an antibody-drug conjugate (ADC) comprised of an anti-EGFR antibody (ABT-806) conjugated to the potent cytotoxic monomethylauristatin F. ABT-414 binds a unique epitope of EGFR which is largely inaccessible when EGFR is expressed at physiological levels but is accessible in tumors that express EGFRde2-7 [also referred to as EGFR variant III (EGFRvIII)] and in tumors with wild-type amplified EGFR or excessive wild-type EGFR activation. These properties of ABT-414 should limit the effects of the toxin on normal tissues. ABT-414 is highly effective in both mutant and wild-type EGFR-positive human tumor xenografts in mice producing complete regressions and cures observed in the most sensitive models. Squamous cell tumors display a relatively high expression of EGFR. Immunohistochemical analysis reveals >70% of squamous lung and esophageal cancers and >90% of oral cavity tumors show moderate to strong staining for EGFR. Given the high unmet need in these patients and the in vivo data supporting improved ABT-414 efficacy in tumors with high EGFR expression, a Phase I/IIa study evaluating the safety, pharmacokinetics and efficacy of ABT-414 in subjects with EGFR-positive, refractory solid tumors has been initiated with a phase IIa expansion at the MTD in squamous NSCLC. Additionally, this study will evaluate the use of SPECT imaging with an indium-111-labeled parental antibody (ABT-806i) as a predictive tool for patient response. Tumor uptake by ABT-806i SPECT imaging correlates with antitumor response in xenograft models, and ABT-806i has been shown to effectively target tumor sites in select cancer patients. A fluorescent in situ hybridization (FISH) probe to detect EGFR amplification and EGFR IHC are also being evaluated with the goal of identifying patients who are most likely to respond to ABT-414 treatment. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A250. Citation Format: Andrew C. Phillips, Erwin R. Boghaert, Kedar S. Vaidya, Peter J. Ansell, David R. Shalinsky, Yumin Zhang, Martin J. Voorbach, Sarah Mudd, Kyle D. Holen, Rod A. Humerickhouse, Edward B. Reilly. ABT-414: An anti-EGFR antibody-drug conjugate as a potential therapeutic for the treatment of patients with squamous cell tumors. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A250.


Molecular Cancer Therapeutics | 2018

Characterization of ABBV-221, a Tumor-Selective EGFR Targeting Antibody Drug Conjugate

Andrew C. Phillips; Erwin R. Boghaert; Kedar S. Vaidya; Hugh D. Falls; Michael J. Mitten; Peter J. DeVries; Lorenzo Benatuil; Chung-Ming Hsieh; Jonathan A. Meulbroek; Sanjay C. Panchal; Fritz G. Buchanan; Kenneth R. Durbin; Martin J. Voorbach; David R. Reuter; Sarah R. Mudd; Lise I. Loberg; Sherry L. Ralston; Diana Cao; Hui K. Gan; Andrew M. Scott; Edward B. Reilly

Depatuxizumab mafodotin (depatux-m, ABT-414) is a tumor-selective antibody drug conjugate (ADC) comprised of the anti-EGFR antibody ABT-806 and the monomethyl auristatin F (MMAF) warhead. Depatux-m has demonstrated promising clinical activity in glioblastoma multiforme (GBM) patients and is currently being evaluated in clinical trials in first-line and recurrent GBM disease settings. Depatux-m responses have been restricted to patients with amplified EGFR, highlighting the need for therapies with activity against tumors with nonamplified EGFR overexpression. In addition, depatux-m dosing has been limited by corneal side effects common to MMAF conjugates. We hypothesized that a monomethyl auristatin E (MMAE) ADC utilizing an EGFR-targeting antibody with increased affinity may have broader utility against tumors with more modest EGFR overexpression while mitigating the risk of corneal side effects. We describe here preclinical characterization of ABBV-221, an EGFR-targeting ADC comprised of an affinity-matured ABT-806 conjugated to MMAE. ABBV-221 binds to a similar EGFR epitope as depatux-m and retains tumor selectivity with increased binding to EGFR-positive tumor cells and greater in vitro potency. ABBV-221 displays increased tumor uptake and antitumor activity against wild-type EGFR-positive xenografts with a greatly reduced incidence of corneal side effects relative to depatux-m. ABBV-221 has similar activity as depatux-m against an EGFR-amplified GBM patient derived xenograft (PDX) model and is highly effective alone and in combination with standard-of-care temozolomide in an EGFRvIII-positive GBM xenograft model. Based on these results, ABBV-221 has advanced to a phase I clinical trial in patients with advanced solid tumors associated with elevated levels of EGFR. Mol Cancer Ther; 17(4); 795–805. ©2018 AACR.


Pharmacology | 2017

A “Prozone-Like” Effect Influences the Efficacy of the Monoclonal Antibody ABT-700 against the Hepatocyte Growth Factor Receptor

Kedar S. Vaidya; Anatol Oleksijew; Lora A. Tucker; William N. Pappano; Mark G. Anderson; Christine M. Grinnell; Qian Zhang; Sarah J. Heighton; Michael J. Mitten; Sasmita Mishra; Joann P. Palma; Jieyi Wang; Edward B. Reilly; Erwin R. Boghaert

ABT-700 is a therapeutic antibody against the hepatocyte growth factor receptor (MET). At doses or regimens that lead to exposures exceeding optimum in vivo, the efficacy of ABT-700 is unexpectedly reduced. We hypothesized that this reduction in efficacy was due to a “prozone-like” effect in vivo. A prozone-like effect, which is a reduction in efficacy beyond optimum exposure, is caused due a mechanism similar to the generation of false negative flocculation tests by excessive antibody titres. In vitro, we demonstrate that at higher ABT-700 concentrations, this “prozone-like” effect is mediated by a progressive conversion from bivalent to ineffective monovalent binding of the antibody. In vivo, the efficacy of ABT-700 is dependent on an optimum range of exposure as well. Our data suggest that the “prozone-like” effect is operative and independent of target expression. ABT-700 dose, regimen, exposure, and tumor burden are interdependent variables influencing the “prozone-like” effect and mediating and in vivo efficacy. By optimization of dosage and regimen we demonstrate that the “prozone-like” effect can be alleviated and ABT-700 efficacy at varying tumor loads can be further extended in combination with cisplatin. Our results suggest that optimization of exposure taking tumor burden into account may alleviate “prozone-like” effects without compromising efficacy.

Collaboration


Dive into the Edward B. Reilly's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susan E. Lacy

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Qian Zhang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Emma Fung

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John E. Harlan

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge