Network


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

Hotspot


Dive into the research topics where Binyam Bezabeh is active.

Publication


Featured researches published by Binyam Bezabeh.


Science Translational Medicine | 2014

A multifunctional bispecific antibody protects against Pseudomonas aeruginosa

Antonio DiGiandomenico; Ashley E. Keller; Cuihua Gao; Godfrey Rainey; Paul Warrener; Mareia M. Camara; Ryan Fleming; Binyam Bezabeh; Nazzareno Dimasi; Bret R. Sellman; Jamese J. Hilliard; Caitlin M. Guenther; Vivekananda Datta; Wei Zhao; Changshou Gao; Xiang-Qing Yu; JoAnn Suzich; C. Kendall Stover

A new antibody platform combining anti-Psl and anti-PcrV activities provides enhanced protection and acts synergistically with antibiotics against Pseudomonas aeruginosa. Bispecific Antibodies Protect Against Pseudomonas Multifunctional bispecific antibodies were constructed conferring three mechanisms of action against the bacterial pathogen Pseudomonas aeruginosa by targeting both the type III secretion injectisome virulence factor PcrV and the persistence factor Psl exopolysaccharide (DiGiandomenico et al.). A new multimechanistic bispecific antibody platform called BiS4αPa exhibited targeted synergistic protection against P. aeruginosa in a mouse model of lung infection compared to the parent monoclonal antibody combination. This BiS4αPa construct, now designated clinical candidate MEDI3902, was also protective in mouse models of thermal injury, bacteremia, and immunosuppression and synergistically enhanced treatment with multiple antibiotic classes. This study suggests that multifunctional bispecific antibodies may be a promising platform for targeting other antibiotic-resistant bacterial pathogens. Widespread drug resistance due to empiric use of broad-spectrum antibiotics has stimulated development of bacteria-specific strategies for prophylaxis and therapy based on modern monoclonal antibody (mAb) technologies. However, single-mechanism mAb approaches have not provided adequate protective activity in the clinic. We constructed multifunctional bispecific antibodies, each conferring three mechanisms of action against the bacterial pathogen Pseudomonas aeruginosa by targeting the serotype-independent type III secretion system (injectisome) virulence factor PcrV and persistence factor Psl exopolysaccharide. A new bispecific antibody platform, BiS4, exhibited superior synergistic protection against P. aeruginosa–induced murine pneumonia compared to parent mAb combinations or other available bispecific antibody structures. BiS4αPa was protective in several mouse infection models against disparate P. aeruginosa strains and unexpectedly further synergized with multiple antibiotic classes even against drug-resistant clinical isolates. In addition to resulting in a multimechanistic clinical candidate (MEDI3902) for the prevention or treatment of P. aeruginosa infections, these antibody studies suggest that multifunctional antibody approaches may be a promising platform for targeting other antibiotic-resistant bacterial pathogens.


Cancer Cell | 2016

A Biparatopic HER2-Targeting Antibody-Drug Conjugate Induces Tumor Regression in Primary Models Refractory to or Ineligible for HER2-Targeted Therapy.

John Y. Li; Samuel R. Perry; Vanessa Muniz-Medina; Xinzhong Wang; Leslie Wetzel; Marlon Rebelatto; Mary Jane Hinrichs; Binyam Bezabeh; Ryan Fleming; Nazzareno Dimasi; Hui Feng; Dorin Toader; Andy Q. Yuan; Lan Xu; Jia Lin; Changshou Gao; Herren Wu; Rakesh Dixit; Jane K. Osbourn; Steven Coats

Antibody-drug conjugate (ADC) which delivers cytotoxic drugs specifically into targeted cells through internalization and lysosomal trafficking has emerged as an effective cancer therapy. We show that a bivalent biparatopic antibody targeting two non-overlapping epitopes on HER2 can induce HER2 receptor clustering, which in turn promotes robust internalization, lysosomal trafficking, and degradation. When conjugated with a tubulysin-based microtubule inhibitor, the biparatopic ADC demonstrates superior anti-tumor activity over ado-trastuzumab emtansine (T-DM1) in tumor models representing various patient subpopulations, including T-DM1 eligible, T-DM1 ineligible, and T-DM1 relapsed/refractory. Our findings indicate that this biparatopic ADC has promising potential as an effective therapy for metastatic breast cancer and a broader patient population may benefit from this unique HER2-targeting ADC.


Journal of Controlled Release | 2015

Stabilization of cysteine-linked antibody drug conjugates with N-aryl maleimides.

R. James Christie; Ryan Fleming; Binyam Bezabeh; Rob Woods; Shenlan Mao; Jay Harper; Augustine Rudolph Joseph; Qianli Wang; Ze-Qi Xu; Herren Wu; Changshou Gao; Nazzareno Dimasi

Maleimides are often used to covalently attach drugs to cysteine thiols for production of antibody-drug conjugates (ADCs). However, ADCs formed with traditional N-alkyl maleimides have variable stability in the bloodstream leading to loss of drug. Here, we report that N-aryl maleimides form stable antibody conjugates under very mild conditions while also maintaining high conjugation efficiency. Thiol-maleimide coupling and ADC stabilization via thiosuccinimide hydrolysis were accelerated by addition of N-phenyl or N-fluorophenyl groups to the ring-head nitrogen. Cysteine-linked ADCs prepared with N-aryl maleimides exhibited less than 20% deconjugation in both thiol-containing buffer and serum when incubated at 37 °C over a period of 7 days, whereas the analogous ADCs prepared with N-alkyl maleimides showed 35-67% deconjugation under the same conditions. ADCs prepared with the anticancer drug N-phenyl maleimide monomethyl-auristatin-E (MMAE) maintained high cytotoxicity following long-term exposure to serum whereas the N-alkyl maleimide MMAE ADC lost potency over time. These data demonstrate that N-aryl maleimides are a convenient and flexible platform to improve the stability of ADCs through manipulation of functional groups attached to the maleimide ring-head nitrogen.


Journal of Controlled Release | 2016

Rational design, biophysical and biological characterization of site-specific antibody-tubulysin conjugates with improved stability, efficacy and pharmacokinetics

Pamela Thompson; Ryan Fleming; Binyam Bezabeh; Fengying Huang; Shenlan Mao; Cui Chen; Jay Harper; Haihong Zhong; Xizhe Gao; Xiang-Qing Yu; Mary Jane Hinrichs; Molly Reed; Adeela Kamal; Patrick Strout; Song Cho; Rob Woods; Robert E. Hollingsworth; Rakesh Dixit; Herren Wu; Changshou Gao; Nazzareno Dimasi

Antibody-drug conjugates (ADCs) are among the most promising empowered biologics for cancer treatment. ADCs are commonly prepared by chemical conjugation of small molecule cytotoxic anti-cancer drugs to antibodies through either lysine side chains or cysteine thiols generated by the reduction of interchain disulfide bonds. Both methods yield heterogeneous conjugates with complex biophysical properties and suboptimal serum stability, efficacy, and pharmacokinetics. To limit the complexity of cysteine-based ADCs, we have engineered and characterized in vitro and in vivo antibody cysteine variants that allow precise control of both site of conjugation and drug load per antibody molecule. We demonstrate that the chemically-defined cysteine-engineered antibody-tubulysin conjugates have improved ex vivo and in vivo stability, efficacy, and pharmacokinetics when compared to conventional cysteine-based ADCs with similar drug-to-antibody ratios. In addition, to limit the non-target FcγRs mediated uptake of the ADCs by cells of the innate immune system, which may result in off-target toxicities, the ADCs have been engineered to lack Fc-receptor binding. The strategies described herein are broadly applicable to any full-length IgG or Fc-based ADC and have been incorporated into an ADC that is in phase I clinical development.


Bioconjugate Chemistry | 2015

Hydrolytically Stable Site-Specific Conjugation at the N-Terminus of an Engineered Antibody

Pamela Thompson; Binyam Bezabeh; Ryan Fleming; Monica Pruitt; Shenlan Mao; Patrick Strout; Cui Chen; Song Cho; Haihong Zhong; Herren Wu; Changshou Gao; Nazzareno Dimasi

Antibody-drug conjugates (ADCs) have emerged as an important class of therapeutics for cancer treatment that combine the target specificity of antibodies with the killing activity of anticancer chemotherapeutics. Early conjugation technologies relied upon random conjugation to either lysine or cysteine residues, resulting in heterogeneous ADCs. Recent technology advancements have resulted in the preparation of homogeneous ADCs through the site-specific conjugation at engineered cysteines, glycosylated amino acids, and bioorthogonal unnatural amino acids. Here we describe for the first time the conjugation of an anti-mitotic drug to an antibody following the mild and selective oxidation of a serine residue engineered at the N-terminus of the light chain. Using an alkoxyamine-derivatized monomethyl auristatine E payload, we have prepared a hydrolytically stable ADC that retains binding to its antigen and displays potent in vitro cytotoxicity and in vivo tumor growth inhibition.


Molecular Pharmaceutics | 2017

Efficient Preparation of Site-Specific Antibody–Drug Conjugates Using Cysteine Insertion

Nazzareno Dimasi; Ryan Fleming; Haihong Zhong; Binyam Bezabeh; Krista Kinneer; Ronald J. Christie; Christine Fazenbaker; Herren Wu; Changshou Gao

Antibody-drug conjugates (ADCs) are a class of biopharmaceuticals that combine the specificity of antibodies with the high-potency of cytotoxic drugs. Engineering cysteine residues in the antibodies using mutagenesis is a common method to prepare site-specific ADCs. With this approach, solvent accessible amino acids in the antibody have been selected for substitution with cysteine for conjugating maleimide-bearing cytotoxic drugs, resulting in homogeneous and stable site-specific ADCs. Here we describe a cysteine engineering approach based on the insertion of cysteines before and after selected sites in the antibody, which can be used for site-specific preparation of ADCs. Cysteine-inserted antibodies have expression level and monomeric content similar to the native antibodies. Conjugation to a pyrrolobenzodiazepine dimer (SG3249) resulted in comparable efficiency of site-specific conjugation between cysteine-inserted and cysteine-substituted antibodies. Cysteine-inserted ADCs were shown to have biophysical properties, FcRn, and antigen binding affinity similar to the cysteine-substituted ADCs. These ADCs were comparable for serum stability to the ADCs prepared using cysteine-mutagenesis and had selective and potent cytotoxicity against human prostate cancer cells. Two of the cysteine-inserted variants abolish binding of the resulting ADCs to FcγRs in vitro, thereby potentially preventing non-target mediated uptake of the ADCs by cells of the innate immune system that express FcγRs, which may result in mitigating off-target toxicities. A selected cysteine-inserted ADC demonstrated potent dose-dependent anti-tumor activity in a xenograph tumor mouse model of human breast adenocarcinoma expressing the oncofetal antigen 5T4.


Cancer Research | 2017

Antibody-drug conjugates bearing pyrrolobenzodiazepine or tubulysin payloads are immunomodulatory and synergize with multiple immunotherapies

Jonathan Rios-Doria; Jay Harper; Raymond Rothstein; Leslie Wetzel; Jon Chesebrough; Allison M. Marrero; Cui Chen; Patrick Strout; Kathy Mulgrew; Kelly McGlinchey; Ryan Fleming; Binyam Bezabeh; John Meekin; David B. Stewart; Maureen Kennedy; Philip Martin; Andrew Buchanan; Nazzareno Dimasi; Emil Michelotti; Robert E. Hollingsworth

Immunogenic cell death (ICD) is the process by which certain cytotoxic drugs induce apoptosis of tumor cells in a manner that stimulates the immune system. In this study, we investigated whether antibody-drug conjugates (ADCS) conjugated with pyrrolobenzodiazepine dimer (PBD) or tubulysin payloads induce ICD, modulate the immune microenvironment, and could combine with immuno-oncology drugs to enhance antitumor activity. We show that these payloads on their own induced an immune response that prevented the growth of tumors following subsequent tumor cell challenge. ADCs had greater antitumor activity in immunocompetent versus immunodeficient mice, demonstrating a contribution of the immune system to the antitumor activity of these ADCs. ADCs also induced immunologic memory. In the CT26 model, depletion of CD8+ T cells abrogated the activity of ADCs when used alone or in combination with a PD-L1 antibody, confirming a role for T cells in antitumor activity. Combinations of ADCs with immuno-oncology drugs, including PD-1 or PD-L1 antibodies, OX40 ligand, or GITR ligand fusion proteins, produced synergistic antitumor responses. Importantly, synergy was observed in some cases with suboptimal doses of ADCs, potentially providing an approach to achieve potent antitumor responses while minimizing ADC-induced toxicity. Immunophenotyping studies in different tumor models revealed broad immunomodulation of lymphoid and myeloid cells by ADC and ADC/immuno-oncology combinations. These results suggest that it may be possible to develop novel combinatorial therapies with PBD- and tubulysin-based ADC and immuno-oncology drugs that may increase clinical responses. Cancer Res; 77(10); 2686-98. ©2017 AACR.


mAbs | 2017

Insertion of scFv into the hinge domain of full-length IgG1 monoclonal antibody results in tetravalent bispecific molecule with robust properties.

Binyam Bezabeh; Ryan Fleming; Christine Fazenbaker; Haihong Zhong; Karen Coffman; Xiang-Qing Yu; Ching Ching Leow; Nerea Gibson; Susan Wilson; C. Kendall Stover; Herren Wu; Changshou Gao; Nazzareno Dimasi

ABSTRACT By simultaneous binding two disease mediators, bispecific antibodies offer the opportunity to broaden the utility of antibody-based therapies. Herein, we describe the design and characterization of Bs4Ab, an innovative and generic bispecific tetravalent antibody platform. The Bs4Ab format comprises a full-length IgG1 monoclonal antibody with a scFv inserted into the hinge domain. The Bs4Ab design demonstrates robust manufacturability as evidenced by MEDI3902, which is currently in clinical development. To further demonstrate the applicability of the Bs4Ab technology, we describe the molecular engineering, biochemical, biophysical, and in vivo characterization of a bispecific tetravalent Bs4Ab that, by simultaneously binding vascular endothelial growth factor and angiopoietin-2, inhibits their function. We also demonstrate that the Bs4Ab platform allows Fc-engineering similar to that achieved with IgG1 antibodies, such as mutations to extend half-life or modulate effector functions.


Clinical Cancer Research | 2017

Fractionated Dosing Improves Preclinical Therapeutic Index of Pyrrolobenzodiazepine-Containing Antibody Drug Conjugates

Mary Jane Hinrichs; Pauline M. Ryan; Bo Zheng; Shameen Afif-Rider; Xiang Qing Yu; Michele Gunsior; Haihong Zhong; Jay Harper; Binyam Bezabeh; Kapil Vashisht; Marlon Rebelatto; Molly Reed; Patricia C. Ryan; Shannon Breen; Neki V. Patel; Cui Chen; Luke Masterson; Arnaud Tiberghien; Phillip W. Howard; Nazzareno Dimasi; Rakesh Dixit

Purpose: To use preclinical models to identify a dosing schedule that improves tolerability of highly potent pyrrolobenzodiazepine dimers (PBDs) antibody drug conjugates (ADCs) without compromising antitumor activity. Experimental Design: A series of dose-fractionation studies were conducted to investigate the pharmacokinetic drivers of safety and efficacy of PBD ADCs in animal models. The exposure–activity relationship was investigated in mouse xenograft models of human prostate cancer, breast cancer, and gastric cancer by comparing antitumor activity after single and fractionated dosing with tumor-targeting ADCs conjugated to SG3249, a potent PBD dimer. The exposure–tolerability relationship was similarly investigated in rat and monkey toxicology studies by comparing tolerability, as assessed by survival, body weight, and organ-specific toxicities, after single and fractionated dosing with ADCs conjugated to SG3249 (rats) or SG3400, a structurally related PBD (monkeys). Results: Observations of similar antitumor activity in mice treated with single or fractionated dosing suggests that antitumor activity of PBD ADCs is more closely related to total exposure (AUC) than peak drug concentrations (Cmax). In contrast, improved survival and reduced toxicity in rats and monkeys treated with a fractionated dosing schedule suggests that tolerability of PBD ADCs is more closely associated with Cmax than AUC. Conclusions: We provide the first evidence that fractionated dosing can improve preclinical tolerability of at least some PBD ADCs without compromising efficacy. These findings suggest that preclinical exploration of dosing schedule could be an important clinical strategy to improve the therapeutic window of highly potent ADCs and should be investigated further. Clin Cancer Res; 23(19); 5858–68. ©2017 AACR.


mAbs | 2017

Guiding bispecific monovalent antibody formation through proteolysis of IgG1 single-chain

Nazzareno Dimasi; Ryan Fleming; Kris Sachsenmeier; Binyam Bezabeh; Carl Hay; Jincheng Wu; Erin Sult; Saravanan Rajan; Li Zhuang; Peter Cariuk; Andrew Buchanan; Michael A. Bowen; Herren Wu; Changshou Gao

ABSTRACT We developed an IgG1 domain-tethering approach to guide the correct assembly of 2 light and 2 heavy chains, derived from 2 different antibodies, to form bispecific monovalent antibodies in IgG1 format. We show here that assembling 2 different light and heavy chains by sequentially connecting them with protease-cleavable polypeptide linkers results in the generation of monovalent bispecific antibodies that have IgG1 sequence, structure and functional properties. This approach was used to generate a bispecific monovalent antibody targeting the epidermal growth factor receptor and the type I insulin-like growth factor receptor that: 1) can be produced and purified using standard IgG1 techniques; 2) exhibits stability and structural features comparable to IgG1; 3) binds both targets simultaneously; and 4) has potent anti-tumor activity. Our strategy provides new engineering opportunities for bispecific antibody applications, and, most importantly, overcomes some of the limitations (e.g., half-antibody and homodimer formation, light chains mispairing, multi-step purification), inherent with some of the previously described IgG1-based bispecific monovalent antibodies.

Collaboration


Dive into the Binyam Bezabeh's collaboration.

Researchain Logo
Decentralizing Knowledge