Network


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

Hotspot


Dive into the research topics where Penelope M. Drake is active.

Publication


Featured researches published by Penelope M. Drake.


Bioconjugate Chemistry | 2014

Aldehyde Tag Coupled with HIPS Chemistry Enables the Production of ADCs Conjugated Site-Specifically to Different Antibody Regions with Distinct in Vivo Efficacy and PK Outcomes

Penelope M. Drake; Aaron E. Albers; Jeanne Baker; Stefanie Bañas; Robyn M. Barfield; Abhijit Bhat; Gregory W. de Hart; Albert W. Garofalo; Patrick G. Holder; Lesley C. Jones; Romas Kudirka; Jesse M. McFarland; Wes Zmolek; David Rabuka

It is becoming increasingly clear that site-specific conjugation offers significant advantages over conventional conjugation chemistries used to make antibody–drug conjugates (ADCs). Site-specific payload placement allows for control over both the drug-to-antibody ratio (DAR) and the conjugation site, both of which play an important role in governing the pharmacokinetics (PK), disposition, and efficacy of the ADC. In addition to the DAR and site of conjugation, linker composition also plays an important role in the properties of an ADC. We have previously reported a novel site-specific conjugation platform comprising linker payloads designed to selectively react with site-specifically engineered aldehyde tags on an antibody backbone. This chemistry results in a stable C–C bond between the antibody and the cytotoxin payload, providing a uniquely stable connection with respect to the other linker chemistries used to generate ADCs. The flexibility and versatility of the aldehyde tag conjugation platform has enabled us to undertake a systematic evaluation of the impact of conjugation site and linker composition on ADC properties. Here, we describe the production and characterization of a panel of ADCs bearing the aldehyde tag at different locations on an IgG1 backbone conjugated using Hydrazino-iso-Pictet-Spengler (HIPS) chemistry. We demonstrate that in a panel of ADCs with aldehyde tags at different locations, the site of conjugation has a dramatic impact on in vivo efficacy and pharmacokinetic behavior in rodents; this advantage translates to an improved safety profile in rats as compared to a conventional lysine conjugate.


Bioconjugate Chemistry | 2013

Hydrazino-Pictet-Spengler ligation as a biocompatible method for the generation of stable protein conjugates.

Paresh Agarwal; Romas Kudirka; Aaron E. Albers; Robyn M. Barfield; Gregory W. de Hart; Penelope M. Drake; Lesley C. Jones; David Rabuka

Aldehyde- and ketone-functionalized biomolecules have found widespread use in biochemical and biotechnological fields. They are typically conjugated with hydrazide or aminooxy nucleophiles under acidic conditions to yield hydrazone or oxime products that are relatively stable, but susceptible to hydrolysis over time. We introduce a new reaction, the hydrazino-Pictet-Spengler (HIPS) ligation, which has two distinct advantages over hydrazone and oxime ligations. First, the HIPS ligation proceeds quickly near neutral pH, allowing for one-step labeling of aldehyde-functionalized proteins under mild conditions. Second, the HIPS ligation product is very stable (>5 days) in human plasma relative to an oxime-linked conjugate (∼1 day), as demonstrated by monitoring protein-fluorophore conjugates by ELISA. Thus, the HIPS ligation exhibits a combination of product stability and speed near neutral pH that is unparalleled by current carbonyl bioconjugation chemistries.


Journal of Biological Chemistry | 2015

Reconstitution of Formylglycine-generating Enzyme with Copper(II) for Aldehyde Tag Conversion

Patrick G. Holder; Lesley C. Jones; Penelope M. Drake; Robyn M. Barfield; Stefanie Bañas; Gregory W. de Hart; Jeanne Baker; David Rabuka

Background: Aerobic formylglycine-generating enzyme (FGE) converts cysteine to formylglycine in vivo. Results: Purified FGE requires preactivation with copper to convert cysteine to formylglycine in vitro. Conclusion: FGE is a metalloenzyme. It is also a useful biocatalyst for the production of proteins that contain aldehyde tags. Significance: Understanding FGE biochemistry informs research on sulfatases and enables expanded biotechnology applications of the aldehyde tag. To further our aim of synthesizing aldehyde-tagged proteins for research and biotechnology applications, we developed methods for recombinant production of aerobic formylglycine-generating enzyme (FGE) in good yield. We then optimized the FGE biocatalytic reaction conditions for conversion of cysteine to formylglycine in aldehyde tags on intact monoclonal antibodies. During the development of these conditions, we discovered that pretreating FGE with copper(II) is required for high turnover rates and yields. After further investigation, we confirmed that both aerobic prokaryotic (Streptomyces coelicolor) and eukaryotic (Homo sapiens) FGEs contain a copper cofactor. The complete kinetic parameters for both forms of FGE are described, along with a proposed mechanism for FGE catalysis that accounts for the copper-dependent activity.


Current Opinion in Chemical Biology | 2015

An emerging playbook for antibody–drug conjugates: lessons from the laboratory and clinic suggest a strategy for improving efficacy and safety

Penelope M. Drake; David Rabuka

Antibody-drug conjugates (ADCs) have become de rigueur for pharmaceutical oncology drug development pipelines. There are more than 40 ADCs undergoing clinical trials and many more in preclinical development. The field has rushed to follow the initial successes of Kadcyla™ and Adcetris™, and moved forward with new targets without much pause for optimization. In some respects, the ADC space has become divided into the clinical realm-where the proven technologies continue to represent the bulk of clinical candidates with a few exceptions-and the research realm-where innovations in conjugation chemistry and linker technologies have suggested that there is much room for improvement in the conventional methods. Now, two and four years after the approvals of Kadcyla™ and Adcetris™, respectively, consensus may at last be building that these two drugs rely on rather unique target antigens that enable their success. It is becoming increasingly clear that future target antigens will require additional innovative approaches. Next-generation ADCs have begun to move out of the lab and into the clinic, where there is a pressing need for continued innovation to overcome the twin challenges of safety and efficacy.


mAbs | 2017

Insights from native mass spectrometry approaches for top- and middle- level characterization of site-specific antibody-drug conjugates

Thomas Botzanowski; Stéphane Erb; Oscar Hernandez-Alba; Anthony Ehkirch; Olivier Colas; Elsa Wagner-Rousset; David Rabuka; Alain Beck; Penelope M. Drake; Sarah Cianférani

ABSTRACT Antibody-drug conjugates (ADCs) have emerged as a family of compounds with promise as efficient immunotherapies. First-generation ADCs were generated mostly via reactions on either lysine side-chain amines or cysteine thiol groups after reduction of the interchain disulfide bonds, resulting in heterogeneous populations with a variable number of drug loads per antibody. To control the position and the number of drug loads, new conjugation strategies aiming at the generation of more homogeneous site-specific conjugates have been developed. We report here the first multi-level characterization of a site-specific ADC by state-of-the-art mass spectrometry (MS) methods, including native MS and its hyphenation to ion mobility (IM-MS). We demonstrate the versatility of native MS methodologies for site-specific ADC analysis, with the unique ability to provide several critical quality attributes within one single run, along with a direct snapshot of ADC homogeneity/heterogeneity without extensive data interpretation. The capabilities of native IM-MS to directly access site-specific ADC conformational information are also highlighted. Finally, the potential of these techniques for assessing an ADCs heterogeneity/homogeneity is illustrated by comparing the analytical characterization of a site-specific DAR4 ADC to that of first-generation ADCs. Altogether, our results highlight the compatibility, versatility, and benefits of native MS approaches for the analytical characterization of all types of ADCs, including site-specific conjugates. Thus, we envision integrating native MS and IM-MS approaches, even in their latest state-of-the-art forms, into workflows that benchmark bioconjugation strategies.


BioDrugs | 2017

Recent Developments in ADC Technology: Preclinical Studies Signal Future Clinical Trends

Penelope M. Drake; David Rabuka

The antibody–drug conjugate (ADC) field is in a transitional period. Older approaches to conjugate composition and dosing regimens still dominate the ADC clinical pipeline, but preclinical work is driving a rapid evolution in how we strategize to improve efficacy and reduce toxicity towards better therapeutic outcomes. These advances are largely based upon a body of investigational studies that together offer a deeper understanding of the absorption, distribution, metabolism, and excretion (ADME) and drug metabolism and pharmacokinetics (DMPK) fates of both the intact conjugate and its small-molecule component. Knowing where the drug goes and how it is processed allows mechanistic connections to be drawn with commonly observed clinical toxicities. The field is also starting to consider ADC interactions with the immune system and potential synergistic therapeutic opportunities therein. In an indication of future directions for the field, antibody conjugates bearing non-cytotoxic small-molecule payloads are being developed to reduce side effects associated with treatment of chronic diseases. ADCs are not a magic bullet to cure disease. However, they will increasingly become valuable therapeutic tools to improve patient outcomes across a variety of indications.


Journal of Chromatography B | 2016

A simple LC/MRM–MS-based method to quantify free linker-payload in antibody-drug conjugate preparations

Wesley Zmolek; Stefanie Bañas; Robyn M. Barfield; David Rabuka; Penelope M. Drake

Antibody-drug conjugates represent a growing class of biologic drugs that use the targeted specificity of an antibody to direct the localization of a small molecule drug, often a cytotoxic payload. After conjugation, antibody-drug conjugate preparations typically retain a residual amount of free (unconjugated) linker-payload. Monitoring this free small molecule drug component is important due to the potential for free payload to mediate unintended (off-target) toxicity. We developed a simple RP-HPLC/MRM-MS-based assay that can be rapidly employed to quantify free linker-payload. The method uses low sample volumes and offers an LLOQ of 10nM with 370pg on column. This analytical approach was used to monitor free linker-payload removal during optimization of the tangential flow filtration manufacturing step.


Archive | 2013

Antibody-Drug Conjugates: Can Coupling Cytotoxicity and Specificity Overcome Therapeutic Resistance?

Penelope M. Drake; David Rabuka

As their name implies, antibody-drug conjugates (ADCs) comprise a humanized or fully human IgG coupled to a cytotoxic small molecule payload. The former component provides the ADC with an antibody’s exquisite selectivity for its target antigen, which is typically a cell surface molecule with a tumor-restricted expression pattern. The latter component provides the ADC with a highly potent cytotoxic payload that can efficiently kill targeted cells at low concentrations (sub nM). As relatively recent additions to the armament of anti-cancer drugs, ADCs hold great promise for disease management, particularly with respect to difficult to treat, drug-resistant tumors. Here, we provide an overview of modern approaches to cancer treatment, from first-generation systemically delivered chemotherapeutics, through the contemporary use of molecularly-targeted small molecules and biologics, to the culmination of both of these approaches—the ADC. Next, we detail the particular features of ADCs that relate to their potential for overcoming drug resistance, namely, the cytotoxic small molecule and the linker component. The latter is exceptionally well-suited to modifications that can render cytotoxic payloads poor substrates for multidrug transporter-mediated efflux from the cell. Finally, we touch on the possibility that ADCs might also serve as an effective means to treat tumors that are resistant to unmodified monoclonal antibody therapies (e.g., rituximab and trastuzumab).


Molecular Cancer Therapeutics | 2018

CAT-02-106, a Site-Specifically Conjugated Anti-CD22 Antibody Bearing an MDR1-Resistant Maytansine Payload Yields Excellent Efficacy and Safety in Preclinical Models

Penelope M. Drake; Adam Carlson; Jesse M. McFarland; Stefanie Bañas; Robyn M. Barfield; Wesley Zmolek; Yun Cheol Kim; Betty C.B. Huang; Romas Kudirka; David Rabuka

Hematologically derived tumors make up ∼10% of all newly diagnosed cancer cases in the United States. Of these, the non-Hodgkin lymphoma (NHL) designation describes a diverse group of cancers that collectively rank among the top 10 most commonly diagnosed cancers worldwide. Although long-term survival trends are improving, there remains a significant unmet clinical need for treatments to help patients with relapsed or refractory disease, one cause of which is drug efflux through upregulation of xenobiotic pumps, such as MDR1. CD22 is a clinically validated target for the treatment of NHL, but no anti-CD22 agents have yet been approved for this indication. Recent approval of an anti-CD22 antibody–drug conjugate (ADC) for the treatment of relapsed/refractory ALL supports the rationale for targeting this protein. An opportunity exists for a next-generation anti-CD22 antibody–drug conjugate (ADC) to address unmet medical needs in the relapsed/refractory NHL population. We describe a site-specifically conjugated antibody–drug conjugate, made using aldehyde tag technology, targeted against CD22 and bearing a noncleavable maytansine payload that is resistant to MDR1-mediated efflux. The construct was efficacious against CD22+ NHL xenografts and could be repeatedly dosed in cynomolgus monkeys at 60 mg/kg with no observed significantly adverse effects. Exposure to total ADC at these doses (as assessed by AUC0-inf) indicated that the exposure needed to achieve efficacy was below tolerable limits. Together, the data suggest that this drug has the potential to be used effectively in patients with CD22+ tumors that have developed MDR1-related resistance to prior therapies. Mol Cancer Ther; 17(1); 161–8. ©2017 AACR.


mAbs | 2018

Antibody-drug conjugate library prepared by scanning insertion of the aldehyde tag into IgG1 constant regions

Betty C.B. Huang; Yun Cheol Kim; Stefanie Bañas; Robyn M. Barfield; Penelope M. Drake; Igor Rupniewski; William E. Haskins; David Rabuka

ABSTRACT The advantages of site-specific over stochastic bioconjugation technologies include homogeneity of product, minimal perturbation of protein structure/function, and – increasingly – the ability to perform structure activity relationship studies at the conjugate level. When selecting the optimal location for site-specific payload placement, many researchers turn to in silico modeling of protein structure to identify regions predicted to offer solvent-exposed conjugatable sites while conserving protein function. Here, using the aldehyde tag as our site-specific technology platform and human IgG1 antibody as our target protein, we demonstrate the power of taking an unbiased scanning approach instead. Scanning insertion of the human formylglycine generating enzyme (FGE) recognition sequence, LCTPSR, at each of the 436 positions in the light and heavy chain antibody constant regions followed by co-expression with FGE yielded a library of antibodies bearing an aldehyde functional group ready for conjugation. Each of the variants was expressed, purified, and conjugated to a cytotoxic payload using the Hydrazinyl Iso-Pictet-Spengler ligation to generate an antibody-drug conjugate (ADC), which was analyzed in terms of conjugatability (assessed by drug-to-antibody ratio, DAR) and percent aggregate. We searched for insertion sites that could generate manufacturable ADCs, defined as those variants yielding reasonable antibody titers, DARs of ≥ 1.3, and ≥ 95% monomeric species. Through this process, we discovered 58 tag insertion sites that met these metrics, including 14 sites in the light chain, a location that had proved refractory to the placement of manufacturable tag sites using in silico modeling/rational approaches.

Collaboration


Dive into the Penelope M. Drake's collaboration.

Top Co-Authors

Avatar

David Rabuka

Catalent Pharma Solutions

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefanie Bañas

Catalent Pharma Solutions

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lesley C. Jones

Catalent Pharma Solutions

View shared research outputs
Top Co-Authors

Avatar

Jeanne Baker

Catalent Pharma Solutions

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge