Network


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

Hotspot


Dive into the research topics where Robert P. Lyon is active.

Publication


Featured researches published by Robert P. Lyon.


Nature Biotechnology | 2014

Self-hydrolyzing maleimides improve the stability and pharmacological properties of antibody-drug conjugates

Robert P. Lyon; Jocelyn R. Setter; Tim D. Bovee; Svetlana O. Doronina; Joshua H. Hunter; Martha Anderson; Cindy Balasubramanian; Steven Duniho; Chris I. Leiske; Fu Li; Peter D. Senter

Many antibody-drug conjugates (ADCs) are unstable in vivo because they are formed from maleimide-containing components conjugated to reactive thiols. These thiosuccinimide linkages undergo two competing reactions in plasma: elimination of the maleimide through a retro-Michael reaction, which results in loss of drug-linker from the ADC, and hydrolysis of the thiosuccinimide ring, which results in a derivative that is resistant to the elimination reaction. In an effort to create linker technologies with improved stability characteristics, we used diaminopropionic acid (DPR) to prepare a drug-linker incorporating a basic amino group adjacent to the maleimide, positioned to provide intramolecular catalysis of thiosuccinimide ring hydrolysis. This basic group induces the thiosuccinimide to undergo rapid hydrolysis at neutral pH and room temperature. Once hydrolyzed, the drug-linker is no longer subject to maleimide elimination reactions, preventing nonspecific deconjugation. In vivo studies demonstrate that the increased stability characteristics can lead to improved ADC antitumor activity and reduced neutropenia.


Bioconjugate Chemistry | 2013

A Potent Anti-CD70 Antibody-Drug Conjugate Combining a Dimeric Pyrrolobenzodiazepine Drug with Site-Specific Conjugation Technology

Scott C. Jeffrey; Patrick J. Burke; Robert P. Lyon; David W. Meyer; Django Sussman; Martha Anderson; Joshua H. Hunter; Chris I. Leiske; Jamie B. Miyamoto; Nicole Nicholas; Nicole M. Okeley; Russell J. Sanderson; Ivan Stone; Weiping Zeng; Stephen J. Gregson; Luke Masterson; Arnaud Tiberghien; Philip W. Howard; David E. Thurston; Che-Leung Law; Peter D. Senter

A highly cytotoxic DNA cross-linking pyrrolobenzodiazepine (PBD) dimer with a valine-alanine dipeptide linker was conjugated to the anti-CD70 h1F6 mAb either through endogenous interchain cysteines or, site-specifically, through engineered cysteines at position 239 of the heavy chains. The h1F6239C-PBD conjugation strategy proved to be superior to interchain cysteine conjugation, affording an antibody-drug conjugate (ADC) with high uniformity in drug-loading and low levels of aggregation. In vitro cytotoxicity experiments demonstrated that the h1F6239C-PBD was potent and immunologically specific on CD70-positive renal cell carcinoma (RCC) and non-Hodgkin lymphoma (NHL) cell lines. The conjugate was resistant to drug loss in plasma and in circulation, and had a pharmacokinetic profile closely matching that of the parental h1F6239C antibody capped with N-ethylmaleimide (NEM). Evaluation in CD70-positive RCC and NHL mouse xenograft models showed pronounced antitumor activities at single or weekly doses as low as 0.1 mg/kg of ADC. The ADC was tolerated at 2.5 mg/kg. These results demonstrate that PBDs can be effectively used for antibody-targeted therapy.


Nature Biotechnology | 2015

Reducing hydrophobicity of homogeneous antibody-drug conjugates improves pharmacokinetics and therapeutic index

Robert P. Lyon; Tim D. Bovee; Svetlana O. Doronina; Patrick J. Burke; Joshua H. Hunter; Haley Neff-LaFord; Mechthild Jonas; Martha Anderson; Jocelyn R. Setter; Peter D. Senter

The in vitro potency of antibody-drug conjugates (ADCs) increases with the drug-to-antibody ratio (DAR); however, ADC plasma clearance also increases with DAR, reducing exposure and in vivo efficacy. Here we show that accelerated clearance arises from ADC hydrophobicity, which can be modulated through drug-linker design. We exemplify this using hydrophilic auristatin drug linkers and PEGylated ADCs that yield uniform, high-DAR ADCs with superior in vivo performance.


Molecular Cancer Therapeutics | 2008

Anti-CD30 diabody-drug conjugates with potent antitumor activity

Kristine M. Kim; Charlotte Mcdonagh; Lori Westendorf; Lindsay L. Brown; Django Sussman; Tiffany Feist; Robert P. Lyon; Stephen C. Alley; Nicole M. Okeley; Xinqun Zhang; Melissa Thompson; Ivan Stone; Hans-Peter Gerber; Paul Carter

Anti-CD30 diabodies were engineered with two cysteine mutations for site-specific drug conjugation in each chain of these homodimeric antibody fragments. Diabodies were conjugated with ∼4 equivalents of the anti-tubulin drugs, monomethyl auristatin E or F, via a protease-cleavable dipeptide linker, to create the conjugates, diabody-vcE4 and diabody-vcF4, respectively. Diabody conjugation had only minor (<3-fold) effects on antigen binding. Diabody-vcF4 was potently cytotoxic against the antigen-positive cell lines, Karpas-299 (34 pmol/L IC50) and L540cy (22 pmol/L IC50), and was 8- and 21-fold more active than diabody-vcE4 against these cell lines, respectively. Clearance of diabody-vcF4 (99-134 mL/d/kg) was 5-fold slower than for the nonconjugated diabody in naive severe combined immunodeficient mice. Diabody-vcF4 had potent and dose-dependent antitumor activity against established Karpas-299 xenografts and gave durable complete responses at well-tolerated doses. Biodistribution experiments with diabody-[3H]-vcF4 (0.72-7.2 mg/kg) in tumor-bearing mice showed a dose-dependent increase in total auristatin accumulation in tumors (≤520 nmol/L) and decrease in relative auristatin accumulation (≤8.1 %ID/g), with peak localization at 4 to 24 h after dosing. Diabody-vcF4 had ∼4-fold lower cytotoxic activity than the corresponding IgG1-vcF4 conjugate in vitro. A similar potency difference was observed in vivo despite 25- to 34-fold faster clearance of diabody-vcF4 than IgG1-vcF4. This may reflect that dose-escalated diabody-vcF4 can surpass IgG1-vcF4 in auristatin delivery to tumors, albeit with higher auristatin exposure to some organs including kidney and liver. Diabody-drug conjugates can have potent antitumor activity at well-tolerated doses and warrant further optimization for cancer therapy. [Mol Cancer Ther 2008;7(8):2486–97]


Molecular Cancer Therapeutics | 2008

Engineered anti-CD70 antibody-drug conjugate with increased therapeutic index

Charlotte Mcdonagh; Kristine M. Kim; Eileen Turcott; Lindsay L. Brown; Lori Westendorf; Tiffany Feist; Django Sussman; Ivan Stone; Martha Anderson; Jamie B. Miyamoto; Robert P. Lyon; Stephen C. Alley; Hans-Peter Gerber; Paul Carter

An anti-CD70 antibody conjugated to monomethylauristatin F (MMAF) via a valine-citrulline dipeptide containing linker has been shown previously to have potent antitumor activity in renal cell cancer xenograft studies. Here, we generated a panel of humanized anti-CD70 antibody IgG variants and conjugated them to MMAF to study the effect of isotype (IgG1, IgG2, and IgG4) and Fcγ receptor binding on antibody-drug conjugate properties. All IgG variants bound CD70+ 786-O cells with an apparent affinity of ∼1 nmol/L, and drug conjugation did not impair antigen binding. The parent anti-CD70 IgG1 bound to human FcγRI and FcγRIIIA V158 and mouse FcγRIV and this binding was not impaired by drug conjugation. In contrast, binding to these Fcγ receptors was greatly reduced or abolished in the variant, IgG1v1, containing the previously described mutations, E233P:L234V:L235A. All conjugates had potent cytotoxic activity against six different antigen-positive cancer cell lines in vitro with IC50 values of 30 to 540 pmol/L. The IgGv1 conjugate with MMAF displayed improved antitumor activity compared with other conjugates in 786-O and UMRC3 models of renal cell cancer and in the DBTRG05-MG glioblastoma model. All conjugates were tolerated to ≥40 mg/kg in mice. Thus, the IgG1v1 MMAF conjugate has an increased therapeutic index compared with the parent IgG1 conjugate. The improved antitumor activity of the IgG1v1 auristatin conjugates may relate to increased exposure as suggested by pharmacokinetic analysis. The strategy used here for enhancing the therapeutic index of antibody-drug conjugates is independent of the antigen-binding variable domains and potentially applicable to other antibodies. [Mol Cancer Ther 2008;7(9):2913–23]


Cancer Research | 2017

Abstract 56: Antibody-drug conjugates containing glucuronide-tubulysin payloads display activity in MDR+ and heterogeneous tumor models

Patrick J. Burke; Joseph Z. Hamilton; Joshua H. Hunter; Julia H. Cochran; Thomas A. Pires; Christopher I. Leiske; Kim K. Emmerton; Peter D. Senter; Robert P. Lyon; Scott C. Jeffrey

While antibody-drug conjugates (ADCs) find increasing application in cancer treatment regimens, de novo or treatment-emergent resistance mechanisms could impair clinical benefit. Two resistance mechanisms that emerge under continuous ADC exposure in vitro include upregulation of transporters that confer multidrug resistance (MDR+) and loss of cognate antigen expression. New technologies that circumvent these resistance mechanisms may serve to extend the utility of next generation ADCs. Recently, we developed the quaternary ammonium linker system to expand the scope of conjugatable payloads to include tertiary amine-containing compounds and applied the linker to tubulysins, a highly potent class of microtubule disrupting agents that maintain activity in MDR+ cell lines. Quaternary ammonium-linked glucuronide-tubulysin drug-linkers were synthesized and evaluated as ADCs. The resulting conjugates were potent and immunologically specific across a panel of cancer cell lines, including those displaying the MDR phenotype. The ADCs also demonstrate potent bystander activity in a co-culture model containing a mixture of antigen-positive and -negative cell lines. Incorporation of a PEG12 side chain in the linker enabled loading at 8-drugs/Ab for increased in vivo potency while maintaining suitable ADC pharmacokinetic properties. In vivo, the glucuronide-tubulysin conjugates displayed activity in MDR+ xenograft models and bystander activity in an admixed Ag+/Ag- heterogeneous tumor model. Thus, the glucuronide-tubulysin drug-linkers represent a promising new payload for ADCs, combining conjugate potency in the presence of the MDR phenotype with robust activity in models of tumor antigen heterogeneity. Citation Format: Patrick J. Burke, Joseph Z. Hamilton, Joshua H. Hunter, Julia H. Cochran, Thomas A. Pires, Christopher I. Leiske, Kim K. Emmerton, Peter D. Senter, Robert P. Lyon, Scott C. Jeffrey. Antibody-drug conjugates containing glucuronide-tubulysin payloads display activity in MDR+ and heterogeneous tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 56. doi:10.1158/1538-7445.AM2017-56


Cancer Research | 2015

Abstract 648: Optimization of a PEGylated glucuronide-auristatin linker for antibody-drug conjugates

Patrick J. Burke; Joseph Z. Hamilton; Scott C. Jeffrey; Joshua H. Hunter; Svetlana O. Doronina; Nicole M. Okeley; Martha Anderson; Peter D. Senter; Robert P. Lyon

The clinical success of antibody-drug conjugates (ADCs) brentuximab vedotin and ado-trastuzumab emtansine has led to an increased effort to identify new antibody payloads and develop improved linker technologies. Most antibody payloads impart significant hydrophobicity to the ADC resulting in accelerated plasma clearance and diminished in vivo activity, particularly for conjugates with high drug to antibody ratios (DAR). Consequently, drug-linkers have been designed to minimize or mask hydrophobicity, enabling a higher DAR and improved pharmacokinetic properties, translating to enhanced in vivo potency. We recently reported on the incorporation of a discrete PEG polymer as a side chain in the β-glucuronide-monomethylauristatin E (MMAE) linker to provide homogeneous DAR 8 conjugates with decreased plasma clearance and increased antitumor activity in xenograft models relative to a non-PEGylated control. The PEG-glucuronide-MMAE lead has now been optimized by minimizing the size of the PEG side chain and incorporating a self-stabilizing maleimide to prevent payload de-conjugation in vivo. Multiple PEG-glucuronide-MMAE linker constructs were prepared with PEG size varying from 0 to 24 ethylene oxide units, and uniform DAR 8 ADCs were evaluated. PEG size had small but variable effects in vitro, with diminished activity observed in some cell lines. In contrast, a clear relationship was observed between PEG length and plasma pharmacokinetics. Longer PEG chains resulted in slower clearance, with a ‘threshold’ length beyond which clearance was not impacted. Conjugates bearing PEG of sufficient length to minimize ADC plasma clearance provided a wider therapeutic window relative to conjugates bearing shorter PEGs. Thus, sufficient masking of drug hydrophobicity is a viable strategy to provide uniform DAR 8 ADCs that preserve ADC pharmacokinetics and increase potency through higher drug loading. Citation Format: Patrick J. Burke, Joseph Z. Hamilton, Scott C. Jeffrey, Joshua H. Hunter, Svetlana O. Doronina, Nicole M. Okeley, Martha E. Anderson, Peter D. Senter, Robert P. Lyon. Optimization of a PEGylated glucuronide-auristatin linker for antibody-drug conjugates. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 648. doi:10.1158/1538-7445.AM2015-648


Cancer Research | 2017

Abstract 4075: Effect of PEG chain length on antibody-drug conjugate tumor and tissue distribution in tumor bearing xenograft mice

Nagendra Chemuturi; Martha Anderson; Julia H. Cochran; Thomas A. Pires; Josh Hunter; David J. Meyer; Jason Neale; Alice Chin; Paul G. Pittman; Cindy Balasubramanian; Francisco Zapata; Jessica K. Simmons; Robert P. Lyon; Steve Alley

Antibody-drug conjugates (ADCs) are a promising new therapeutic modality having demonstrated clinical efficacy in the treatment of cancer. While ADCs have better exposures than their constitutive chemotypes, it is important that they maintain the favorable pharmacokinetics of their antibody backbones. Previous work incorporating PEG into the drug-linker as a sidechain helped to mask the hydrophobicity of the chemotypes, resulting in improved ADC exposures in rats compared to ADCs with non-PEGylated drug-linkers. The current studies were aimed at understanding the effect of PEG chain length on ADC efficacy, tumor and tissue distribution in tumor bearing mice. Efficacy and biodistribution of radiolabeled ADCs with 2, 4, 8, 12 and 24 PEG units in the drug-linker as a side chain were investigated in SCID mice bearing a subcutaneous xenograft of L540cy tumor cells. A non-PEGylated ADC was used as control. Antibody based radioactivity in plasma, tumor, liver, lung, kidney, spleen and brain were measured over 14 days. MMAE, the free drug released from the ADC, was measured by mass spectrometry. Reduction in tumor weight was determined as a measure of efficacy. Compartmental analysis of the plasma exposure data was conducted to understand the change in rate constants with change in PEG chain length. ADCs with side chain PEGylated drug-linkers had greater plasma and tumor exposures than the non-PEGylated control ADC. Increasing PEG chain length in the linker led to increased plasma and tumor exposures, and lower plasma clearances. Tumor distribution was binary with ADCs with 2 and 4 PEG units in the linker showing similar tumor exposures while the ADCs with 8, 12 and 24 PEG units providing similar but significantly higher tumor exposures. ADCs with 8, 12 and 24 PEG units in the linker had significantly higher tumor to plasma exposure ratios than ADCs with 2 and 4-PEG units in the drug-linker. The reduction in tumor weights was also binary. ADCs with 2 and 4 PEG units provided a 35-45% decrease in tumor weights while ADCs with 8, 12 and 24 PEG units in the linker provided 75-85% reduction in tumor weights. The non-PEGylated control ADC decreased the tumor weights by 11%. In general, PEGylated ADCs increased the extent of tissue distribution, but the tissue to plasma ratios remained the same indicating that the increased tissue exposures was a function of increased plasma exposures. However, PEGylated ADC tumor exposure increased beyond that expected based solely on the increase in plasma exposure. Incorporation of PEG as a side chain into the drug-linker improved the tumor disposition of ADCs with little alteration in tissue disposition. ADCs with 8, 12 and 24 PEG units in the drug- linker showed preferential uptake into tumor, but not the tissues as evidenced by tumor (or tissue) to plasma exposure ratios. Over all, the ADCs with 8, 12 and 24 PEG units in the linker provided the optimal combination of tumor distribution, efficacy and tissue distribution. Citation Format: Nagendra V. Chemuturi, Martha Anderson, Julia Cochran, Thomas Pires, Josh Hunter, David Meyer, Jason Neale, Alice Chin, Paul Pittman, Cindy Balasubramanian, Francisco Zapata, Jessica Simmons, Robert Lyon, Steve Alley. Effect of PEG chain length on antibody-drug conjugate tumor and tissue distribution in tumor bearing xenograft mice [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4075. doi:10.1158/1538-7445.AM2017-4075


Journal of the American Chemical Society | 2001

Self-assembly and gelation of oxidized glutathione in organic solvents

Robert P. Lyon; William M. Atkins


Biochemistry | 2003

Novel class of bivalent glutathione S-transferase inhibitors.

Robert P. Lyon; John J. Hill; William M. Atkins

Collaboration


Dive into the Robert P. Lyon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge