51
|
Lee DS, Suh MI, Kang SY, Hwang DW. Physiologic constraints of using exosomes in vivo as systemic delivery vehicles. PRECISION NANOMEDICINE 2019. [DOI: 10.33218/prnano2(3)070819.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Systemic delivery of exosomes meets hurdles which had not been elucidated using live molecular imaging for their biodistribution. Production and uptake of endogenous exosomes are expected to be nonspecific and specific, respectively, where external stimuli of production of exosomes and their quantitative degree of productions are not understood. Despite this lack of understanding of basic physiology of in vivo behavior of exosomes including their possible paracrine or endocrine actions, many engineering efforts are taken to develop therapeutic vehicles. Especially, the fraction of exosomes’ taking the routes of waste disposal and exerting target actions are not characterized after systemic administration. Here, we reviewed the literature about in vivo distribution and disposal/excretion of exogenous or endogenous exosomes and, from these limited resources of knowledge currently available, summarized the knowledge and the uncertainties of exosomes on physiologic standpoints. An eloquent example of the investigations to understand the roles and confounders of exosomes’ action in the brain was highlighted with emphasis on the recent discovery of brain lymphatics and hypothesis of glymphatic/lymphatic clearance pathways in diseases as well as in physiologic processes. The possibility of delivering therapeutic exosomes through the systemic circulation, across blood-brain barriers and finally to target cells such as microglia, astrocytes and/or neurons is a good testbed in which the investigators can formulate problems to solve for both understanding (science) and application (engineering).
Collapse
Affiliation(s)
- Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - MInseok Suh
- 2Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul National University,
| | - Seo Young Kang
- Department of Nuclear Medicine, Ewha Womans University Medical Center, Seoul,
| | - Do Won Hwang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
52
|
Mortensen MR, Skovsgaard MB, Gothelf KV. Considerations on Probe Design for Affinity‐Guided Protein Conjugation. Chembiochem 2019; 20:2711-2728. [DOI: 10.1002/cbic.201900157] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Michael R. Mortensen
- Center for Multifunctional Biomolecular Drug DesignInterdisciplinary Nanoscience CenterAarhus University Gustav Wieds Vej 14 8000 Aarhus C Denmark
- Department of ChemistryAarhus University Langelandsgade 140 8000 Aarhus C Denmark
| | - Mikkel B. Skovsgaard
- Center for Multifunctional Biomolecular Drug DesignInterdisciplinary Nanoscience CenterAarhus University Gustav Wieds Vej 14 8000 Aarhus C Denmark
- Department of ChemistryAarhus University Langelandsgade 140 8000 Aarhus C Denmark
| | - Kurt V. Gothelf
- Center for Multifunctional Biomolecular Drug DesignInterdisciplinary Nanoscience CenterAarhus University Gustav Wieds Vej 14 8000 Aarhus C Denmark
- Department of ChemistryAarhus University Langelandsgade 140 8000 Aarhus C Denmark
| |
Collapse
|
53
|
O'Donoghue JA, Danila DC, Pandit-Taskar N, Beylergil V, Cheal SM, Fleming SE, Fox JJ, Ruan S, Zanzonico PB, Ragupathi G, Lyashchenko SK, Williams SP, Scher HI, Fine BM, Humm JL, Larson SM, Morris MJ, Carrasquillo JA. Pharmacokinetics and Biodistribution of a [ 89Zr]Zr-DFO-MSTP2109A Anti-STEAP1 Antibody in Metastatic Castration-Resistant Prostate Cancer Patients. Mol Pharm 2019; 16:3083-3090. [PMID: 31117485 DOI: 10.1021/acs.molpharmaceut.9b00326] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A six-transmembrane epithelial antigen of prostate-1 (STEAP1) is a newly identified target in prostate cancer. The use of radio-labeled STEAP1-targeting antibodies with positron emission tomography (PET) may allow for detection of sites of metastatic prostate cancer and may refine patient selection for antigen-directed therapies. This was a prospective study in seven patients with metastatic castration-resistant prostate cancer who had at least one archival biopsy that was STEAP1-positive by immunohistochemistry. Patients received intravenous injections of ∼185 MBq and 10 mg of [89Zr]Zr-DFO-MSTP2109A, a humanized IgG1 monoclonal antibody directed against STEAP1. PET/CT images, blood samples, and whole-body counts were monitored longitudinally in six patients. Here, we report on safety, biodistribution, pharmacokinetics, dose estimates to normal tissues, and initial tumor targeting for this group of patients. There was no significant acute or subacute toxicity. Favorable biodistribution and enhanced lesion uptake (in both bone and soft tissue) were observed on imaging using a mass of 10 mg of DFO-MSTP2109A. The best lesion discrimination was seen at the latest imaging time, a median of 6 days postadministration. Pharmacokinetics showed a median serum T1/2 β of 198 h, volume of central compartment of 3.54 L (similar to plasma volume), and clearance of 19.7 mL/h. The median biologic T1/2 for whole-body retention was 469 h. The highest mean absorbed doses to normal organs (mGy/MBq) were 1.18, 1.11, 0.78, 0.73, and 0.71 for liver, heart wall, lung, kidney, and spleen, respectively. Excellent targeting of metastatic prostate sites in both bone and soft tissue was observed, with an optimal imaging time of 6 days postadministration. The liver and heart were the normal organs that experienced the highest absorbed doses. The pharmacokinetics were similar to other antibodies without major cross-reactivity with normal tissues. A more detailed analysis of lesion targeting in a larger patient population with correlation to immunohistology and standard imaging modalities has been reported.
Collapse
Affiliation(s)
| | - Daniel C Danila
- Department of Medicine , Joan and Sanford I. Weill College of Medicine of Cornell University , New York , New York 10065 , United States
| | - Neeta Pandit-Taskar
- Department of Radiology , Joan and Sanford I. Weill Cornell Medical Center , New York , New York 10065 , United States
| | | | | | | | | | | | | | | | | | - Simon P Williams
- Genentech , South San Francisco , California 94080 , United States
| | - Howard I Scher
- Department of Medicine , Joan and Sanford I. Weill College of Medicine of Cornell University , New York , New York 10065 , United States
| | - Bernard M Fine
- Genentech , South San Francisco , California 94080 , United States
| | | | - Steven M Larson
- Department of Radiology , Joan and Sanford I. Weill Cornell Medical Center , New York , New York 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis of the Ludwig Center for Cancer Immunotherapy , New York , New York 10065 , United States
| | - Michael J Morris
- Department of Medicine , Joan and Sanford I. Weill College of Medicine of Cornell University , New York , New York 10065 , United States
| | - Jorge A Carrasquillo
- Department of Radiology , Joan and Sanford I. Weill Cornell Medical Center , New York , New York 10065 , United States.,Center for Targeted Radioimmunotherapy and Diagnosis of the Ludwig Center for Cancer Immunotherapy , New York , New York 10065 , United States
| |
Collapse
|
54
|
Murray JK, Wu B, Tegley CM, Nixey TE, Falsey JR, Herberich B, Yin L, Sham K, Long J, Aral J, Cheng Y, Netirojjanakul C, Doherty L, Glaus C, Ikotun T, Li H, Tran L, Soto M, Salimi-Moosavi H, Ligutti J, Amagasu S, Andrews KL, Be X, Lin MHJ, Foti RS, Ilch CP, Youngblood B, Kornecook TJ, Karow M, Walker KW, Moyer BD, Biswas K, Miranda LP. Engineering Na V1.7 Inhibitory JzTx-V Peptides with a Potency and Basicity Profile Suitable for Antibody Conjugation To Enhance Pharmacokinetics. ACS Chem Biol 2019; 14:806-818. [PMID: 30875193 DOI: 10.1021/acschembio.9b00183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drug discovery research on new pain targets with human genetic validation, including the voltage-gated sodium channel NaV1.7, is being pursued to address the unmet medical need with respect to chronic pain and the rising opioid epidemic. As part of early research efforts on this front, we have previously developed NaV1.7 inhibitory peptide-antibody conjugates with tarantula venom-derived GpTx-1 toxin peptides with an extended half-life (80 h) in rodents but only moderate in vitro activity (hNaV1.7 IC50 = 250 nM) and without in vivo activity. We identified the more potent peptide JzTx-V from our natural peptide collection and improved its selectivity against other sodium channel isoforms through positional analogueing. Here we report utilization of the JzTx-V scaffold in a peptide-antibody conjugate and architectural variations in the linker, peptide loading, and antibody attachment site. We found conjugates with 100-fold improved in vitro potency relative to those of complementary GpTx-1 analogues, but pharmacokinetic and bioimaging analyses of these JzTx-V conjugates revealed a shorter than expected plasma half-life in vivo with accumulation in the liver. In an attempt to increase circulatory serum levels, we sought the reduction of the net +6 charge of the JzTx-V scaffold while retaining a desirable NaV in vitro activity profile. The conjugate of a JzTx-V peptide analogue with a +2 formal charge maintained NaV1.7 potency with 18-fold improved plasma exposure in rodents. Balancing the loss of peptide and conjugate potency associated with the reduction of net charge necessary for improved target exposure resulted in a compound with moderate activity in a NaV1.7-dependent pharmacodynamic model but requires further optimization to identify a conjugate that can fully engage NaV1.7 in vivo.
Collapse
|
55
|
Davydova M, Dewaele Le Roi G, Adumeau P, Zeglis BM. Synthesis and Bioconjugation of Thiol-Reactive Reagents for the Creation of Site-Selectively Modified Immunoconjugates. J Vis Exp 2019. [PMID: 30907883 DOI: 10.3791/59063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Maleimide-bearing bifunctional probes have been employed for decades for the site-selective modification of thiols in biomolecules, especially antibodies. Yet maleimide-based conjugates display limited stability in vivo because the succinimidyl thioether linkage can undergo a retro-Michael reaction. This, of course, can lead to the release of the radioactive payload or its exchange with thiol-bearing biomolecules in circulation. Both of these processes can produce elevated activity concentrations in healthy organs as well as decreased activity concentrations in target tissues, resulting in reduced imaging contrast and lower therapeutic ratios. In 2018, we reported the creation of a modular, stable, and easily accessible phenyloxadiazolyl methyl sulfone reagent - dubbed 'PODS' - as a platform for thiol-based bioconjugations. We have clearly demonstrated that PODS-based site-selective bioconjugations reproducibly and robustly create homogenous, well-defined, highly immunoreactive, and highly stable radioimmunoconjugates. Furthermore, preclinical experiments in murine models of colorectal cancer have shown that these site-selectively labeled radioimmunoconjugates exhibit far superior in vivo performance compared to radiolabeled antibodies synthesized via maleimide-based conjugations. In this protocol, we will describe the four-step synthesis of PODS, the creation of a bifunctional PODS-bearing variant of the ubiquitous chelator DOTA (PODS-DOTA), and the conjugation of PODS-DOTA to the HER2-targeting antibody trastuzumab.
Collapse
Affiliation(s)
- Maria Davydova
- Department of Chemistry, Hunter College of the City University of New York
| | - Guillaume Dewaele Le Roi
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York; Department of Radiology, Memorial Sloan Kettering Cancer Center; Department of Radiology, Weill Cornell Medical College;
| |
Collapse
|
56
|
White JB, Fleming R, Masterson L, Ruddle BT, Zhong H, Fazenbaker C, Strout P, Rosenthal K, Reed M, Muniz-Medina V, Howard P, Dixit R, Wu H, Hinrichs MJ, Gao C, Dimasi N. Design and characterization of homogenous antibody-drug conjugates with a drug-to-antibody ratio of one prepared using an engineered antibody and a dual-maleimide pyrrolobenzodiazepine dimer. MAbs 2019; 11:500-515. [PMID: 30835621 DOI: 10.1080/19420862.2019.1578611] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Most strategies used to prepare homogeneous site-specific antibody-drug conjugates (ADCs) result in ADCs with a drug-to-antibody ratio (DAR) of two. Here, we report a disulfide re-bridging strategy to prepare homogeneous ADCs with DAR of one using a dual-maleimide pyrrolobenzodiazepine (PBD) dimer (SG3710) and an engineered antibody (Flexmab), which has only one intrachain disulfide bridge at the hinge. We demonstrate that SG3710 efficiently re-bridge a Flexmab targeting human epidermal growth factor receptor 2 (HER2), and the resulting ADC was highly resistant to payload loss in serum and exhibited potent anti-tumor activity in a HER2-positive gastric carcinoma xenograft model. Moreover, this ADC was tolerated in rats at twice the dose compared to a site-specific ADC with DAR of two prepared using a single-maleimide PBD dimer (SG3249). Flexmab technologies, in combination with SG3710, provide a platform for generating site-specific homogenous PBD-based ADCs with DAR of one, which have improved biophysical properties and tolerability compared to conventional site-specific PBD-based ADCs with DAR of two.
Collapse
Affiliation(s)
- Jason B White
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Ryan Fleming
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | | | - Ben T Ruddle
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Haihong Zhong
- c Oncology Research , MedImmune , Gaithersburg , MD , USA
| | | | - Patrick Strout
- c Oncology Research , MedImmune , Gaithersburg , MD , USA
| | - Kim Rosenthal
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Molly Reed
- d Biologics Safety Assessment , MedImmune , Gaithersburg , MD , USA
| | | | - Philip Howard
- b Spirogen Ltd , QMB Innovation Center , London , UK
| | - Rakesh Dixit
- d Biologics Safety Assessment , MedImmune , Gaithersburg , MD , USA
| | - Herren Wu
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | | | - Changshou Gao
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| | - Nazzareno Dimasi
- a Antibody Discovery and Protein Engineering , MedImmune , Gaithersburg , MD , USA
| |
Collapse
|
57
|
Chen TH, Yang Y, Zhang Z, Fu C, Zhang Q, Williams JD, Wirth MJ. Native Reversed-Phase Liquid Chromatography: A Technique for LCMS of Intact Antibody-Drug Conjugates. Anal Chem 2019; 91:2805-2812. [PMID: 30661356 PMCID: PMC6727645 DOI: 10.1021/acs.analchem.8b04699] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The synthesis of antibody-drug conjugates (ADCs) using the interchain cysteines of the antibody inherently gives a mixture of proteins with varying drug-to-antibody ratio. The drug distribution profiles of ADCs are routinely characterized by hydrophobic interaction chromatography (HIC). Because HIC is not in-line compatible with mass spectrometry (MS) due to the high salt levels, it is laborious to identify the constituents of HIC peaks. An MS-compatible alternative to HIC is reported here: native reversed phase liquid chromatography (nRPLC). This novel technique employs a mobile phase 50 mM ammonium acetate for high sensitivity in MS and elution with a gradient of water/isopropanol. The key to the enhancement is a bonded phase giving weaker drug-surface interactions compared to the noncovalent interactions holding the antibody-drug conjugates together. The hydrophobicity of the bonded phase is varied, and the least hydrophobic bonded phase in the series, poly(methyl methacrylate), is found to resolve the intact constituents of a model ADC (Ab095-PZ) and a commercial ADC (brentuximab vedotin) under the MS-compatible conditions. The nRPLC-MS data show that all species, ranging from drug-to-antibody ratios of 1 to 8, remained intact in the column. Another desired advantage of the nRPLC is the ability of resolving multiple positional isomers of ADC that are not well-resolved in other chromatographic modes. This supports the premise that lower hydrophobicity of the bonded phase is the key to enabling online nRPLC-MS analysis of antibody-drug conjugates.
Collapse
Affiliation(s)
- Tse-Hong Chen
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Yun Yang
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Zhaorui Zhang
- Process Analytical Chemistry, AbbVie, Inc. 1 N. Waukegan Road, North Chicago, Illinois 60064, United States
| | - Cexiong Fu
- Process Analytical Chemistry, AbbVie, Inc. 1 N. Waukegan Road, North Chicago, Illinois 60064, United States
| | - Qunying Zhang
- Process Analytical Chemistry, AbbVie, Inc. 1 N. Waukegan Road, North Chicago, Illinois 60064, United States
| | - Jon D. Williams
- Discovery Structural Chemistry, AbbVie, Inc. 1 N. Waukegan Road, North Chicago, Illinois 60064, United States
| | - Mary J. Wirth
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
58
|
Bumbaca B, Li Z, Shah DK. Pharmacokinetics of protein and peptide conjugates. Drug Metab Pharmacokinet 2019; 34:42-54. [PMID: 30573392 PMCID: PMC6378135 DOI: 10.1016/j.dmpk.2018.11.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/29/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Abstract
Protein and peptide conjugates have become an important component of therapeutic and diagnostic medicine. These conjugates are primarily designed to improve pharmacokinetics (PK) of those therapeutic or imaging agents, which do not possess optimal disposition characteristics. In this review we have summarized preclinical and clinical PK of diverse protein and peptide conjugates, and have showcased how different conjugation approaches are used to obtain the desired PK. We have classified the conjugates into peptide conjugates, non-targeted protein conjugates, and targeted protein conjugates, and have highlighted diagnostic and therapeutic applications of these conjugates. In general, peptide conjugates demonstrate very short half-life and rapid renal elimination, and they are mainly designed to achieve high contrast ratio for imaging agents or to deliver therapeutic agents at sites not reachable by bulky or non-targeted proteins. Conjugates made from non-targeted proteins like albumin are designed to increase the half-life of rapidly eliminating therapeutic or imaging agents, and improve their delivery to tissues like solid tumors and inflamed joints. Targeted protein conjugates are mainly developed from antibodies, antibody derivatives, or endogenous proteins, and they are designed to improve the contrast ratio of imaging agents or therapeutic index of therapeutic agents, by enhancing their delivery to the site-of-action.
Collapse
Affiliation(s)
- Brandon Bumbaca
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, USA.
| |
Collapse
|
59
|
Morais M, Forte N, Chudasama V, Baker JR. Application of Next-Generation Maleimides (NGMs) to Site-Selective Antibody Conjugation. Methods Mol Biol 2019; 2033:15-24. [PMID: 31332744 DOI: 10.1007/978-1-4939-9654-4_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Site-selective antibody conjugation is widely recognized as a key strategy for the optimum construction of antibody-drug conjugates (ADCs). Achieving such bioconjugation directly onto native antibodies would represent the ideal solution, as it would afford greatly improved homogeneity whilst avoiding the need for genetic engineering, and even allow the repurposing of existing antibodies "off-the shelf." Here we describe a protocol for the use of next-generation maleimides (NGMs) for the selective modification of the four interchain disulfide bonds present in a typical IgG1 antibody format. These reagents retain the efficiency of classical maleimides whilst serving to rebridge each reduced disulfide bond, affording one attachment per disulfide. The approach is simple, uses readily available reagents, and generates robustly stable conjugates which are ideal for in vitro or in vivo applications. In addition to use in the construction of ADCs these reagents can also be used to develop antibody conjugates for imaging, bispecifics, and broadly for use across biology and medicine.
Collapse
Affiliation(s)
- Maurício Morais
- Department of Chemistry, University College London, London, UK
| | - Nafsika Forte
- Department of Chemistry, University College London, London, UK
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, UK
| | - James R Baker
- Department of Chemistry, University College London, London, UK.
| |
Collapse
|
60
|
Cohen DT, Zhang C, Fadzen CM, Mijalis AJ, Hie L, Johnson KD, Shriver Z, Plante O, Miller SJ, Buchwald SL, Pentelute BL. A chemoselective strategy for late-stage functionalization of complex small molecules with polypeptides and proteins. Nat Chem 2019; 11:78-85. [PMID: 30397320 PMCID: PMC6454892 DOI: 10.1038/s41557-018-0154-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 09/07/2018] [Indexed: 01/28/2023]
Abstract
Conjugates between proteins and small molecules enable access to a vast chemical space that is not achievable with either type of molecule alone; however, the paucity of specific reactions capable of functionalizing proteins and natural products presents a formidable challenge for preparing conjugates. Here we report a strategy for conjugating electron-rich (hetero)arenes to polypeptides and proteins. Our bioconjugation technique exploits the electrophilic reactivity of an oxidized selenocysteine residue in polypeptides and proteins, and the electron-rich character of certain small molecules to provide bioconjugates in excellent yields under mild conditions. This conjugation chemistry enabled the synthesis of peptide-vancomycin conjugates without the prefunctionalization of vancomycin. These conjugates have an enhanced in vitro potency for resistant Gram-positive and Gram-negative pathogens. Additionally, we show that a 6 kDa affibody protein and a 150 kDa immunoglobulin-G antibody could be modified without diminishing bioactivity.
Collapse
Affiliation(s)
- Daniel T Cohen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- AbbVie, Inc., North Chicago, IL, USA.
| | - Chi Zhang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Colin M Fadzen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexander J Mijalis
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Liana Hie
- Department of Chemistry, Yale University, New Haven, CT, USA
| | | | | | | | - Scott J Miller
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Stephen L Buchwald
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bradley L Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute, Broad Institute of Harvard and MIT, Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
61
|
Lucas AT, Robinson R, Schorzman AN, Piscitelli JA, Razo JF, Zamboni WC. Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients. Antibodies (Basel) 2019; 8:E3. [PMID: 31544809 PMCID: PMC6640706 DOI: 10.3390/antib8010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/11/2022] Open
Abstract
The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution. This review provides an updated summary of factors known to affect the disposition of mAbs/ADCs in development and in clinical use, as well as how these factors should be considered in the selection and design of preclinical studies of ADC agents in development.
Collapse
Affiliation(s)
- Andrew T Lucas
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Ryan Robinson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Joseph A Piscitelli
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - Juan F Razo
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
62
|
Xie H, Adjei AA. Antibody-Drug Conjugates for the Therapy of Thoracic Malignancies. J Thorac Oncol 2018; 14:358-376. [PMID: 30599202 DOI: 10.1016/j.jtho.2018.11.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/25/2018] [Accepted: 11/29/2018] [Indexed: 10/27/2022]
Abstract
Antibody-drug conjugates (ADCs) are a novel class of therapeutic agents incorporating both target-specific monoclonal antibodies and cytotoxic small molecules via a chemical linker. They were first introduced into the clinic for the treatment of advanced hematologic malignancies. The only approved ADC for solid tumors targets erb-b2 receptor tyrosine kinase (HER2), a validated antigen in breast cancer. Many ADCs are under active investigation for various types of solid tumors. In this article, we review the literature from several perspectives including the design, pharmacology, and mechanism-based toxicities of antibody-drug conjugates. We then discuss ADCs currently in clinical development for thoracic malignancies.
Collapse
Affiliation(s)
- Hao Xie
- Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
63
|
Morais M, Ma MT. Site-specific chelator-antibody conjugation for PET and SPECT imaging with radiometals. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:91-104. [PMID: 30553525 PMCID: PMC6291455 DOI: 10.1016/j.ddtec.2018.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 11/17/2022]
Abstract
Antibodies and their derivatives radiolabelled with positron- and gamma-emitting radiometals enable sensitive and quantitative molecular Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) imaging of antibody distribution in vivo. Chelators that are covalently attached to antibodies allow radiolabelling with metallic PET and SPECT radioisotopes. Conventional strategies for chelator-protein conjugation generate heterogeneous mixtures of bioconjugates that can exhibit reduced affinity for their receptor targets, and undesirable biodistribution and pharmacokinetics. Recent advances in bioconjugation technology enable site-specific modification to generate well-defined constructs with superior properties. Herein we survey existing site-specific chelator-protein conjugation methods. These include chelator attachment to cysteines/disulfide bonds or the glycan region of the antibody, enzyme-mediated chelator conjugation, and incorporation of sequences of amino acids that chelate the radiometal. Such technology will allow better use of PET and SPECT imaging in the development of antibody-based therapies.
Collapse
Affiliation(s)
- Mauricio Morais
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom.
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
64
|
Su Y, Liu Y, Behrens CR, Bidlingmaier S, Lee NK, Aggarwal R, Sherbenou DW, Burlingame AL, Hann BC, Simko JP, Premasekharan G, Paris PL, Shuman MA, Seo Y, Small EJ, Liu B. Targeting CD46 for both adenocarcinoma and neuroendocrine prostate cancer. JCI Insight 2018; 3:121497. [PMID: 30185663 DOI: 10.1172/jci.insight.121497] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022] Open
Abstract
Although initially responsive to androgen signaling inhibitors (ASIs), metastatic castration-resistant prostate cancer (mCRPC) inevitably develops and is incurable. In addition to adenocarcinoma (adeno), neuroendocrine prostate cancer (NEPC) emerges to confer ASI resistance. We have previously combined laser capture microdissection and phage antibody display library selection on human cancer specimens and identified novel internalizing antibodies binding to tumor cells residing in their tissue microenvironment. We identified the target antigen for one of these antibodies as CD46, a multifunctional protein that is best known for negatively regulating the innate immune system. CD46 is overexpressed in primary tumor tissue and CRPC (localized and metastatic; adeno and NEPC), but expressed at low levels on normal tissues except for placental trophoblasts and prostate epithelium. Abiraterone- and enzalutamide-treated mCRPC cells upregulate cell surface CD46 expression. Genomic analysis showed that the CD46 gene is gained in 45% abiraterone-resistant mCRPC patients. We conjugated a tubulin inhibitor to our macropinocytosing anti-CD46 antibody and showed that the resulting antibody-drug conjugate (ADC) potently and selectively kills both adeno and NEPC cell lines in vitro (sub-nM EC50) but not normal cells. CD46 ADC regressed and eliminated an mCRPC cell line xenograft in vivo in both subcutaneous and intrafemoral models. Exploratory toxicology studies of the CD46 ADC in non-human primates demonstrated an acceptable safety profile. Thus, CD46 is an excellent target for antibody-based therapy development, which has potential to be applicable to both adenocarcinoma and neuroendocrine types of mCRPC that are resistant to current treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Rahul Aggarwal
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center
| | | | | | | | - Jeffry P Simko
- Helen Diller Family Comprehensive Cancer Center.,Department of Pathology
| | | | - Pamela L Paris
- Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | | | - Youngho Seo
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California, USA
| | - Eric J Small
- Department of Medicine.,Helen Diller Family Comprehensive Cancer Center.,Department of Urology, and
| | - Bin Liu
- Department of Anesthesia.,Helen Diller Family Comprehensive Cancer Center
| |
Collapse
|
65
|
Figueroa I, Leipold D, Leong S, Zheng B, Triguero-Carrasco M, Fourie-O'Donohue A, Kozak KR, Xu K, Schutten M, Wang H, Polson AG, Kamath AV. Prediction of non-linear pharmacokinetics in humans of an antibody-drug conjugate (ADC) when evaluation of higher doses in animals is limited by tolerability: Case study with an anti-CD33 ADC. MAbs 2018; 10:738-750. [PMID: 29757698 PMCID: PMC6150628 DOI: 10.1080/19420862.2018.1465160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/03/2018] [Accepted: 04/09/2018] [Indexed: 11/01/2022] Open
Abstract
For antibody-drug conjugates (ADCs) that carry a cytotoxic drug, doses that can be administered in preclinical studies are typically limited by tolerability, leading to a narrow dose range that can be tested. For molecules with non-linear pharmacokinetics (PK), this limited dose range may be insufficient to fully characterize the PK of the ADC and limits translation to humans. Mathematical PK models are frequently used for molecule selection during preclinical drug development and for translational predictions to guide clinical study design. Here, we present a practical approach that uses limited PK and receptor occupancy (RO) data of the corresponding unconjugated antibody to predict ADC PK when conjugation does not alter the non-specific clearance or the antibody-target interaction. We used a 2-compartment model incorporating non-specific and specific (target mediated) clearances, where the latter is a function of RO, to describe the PK of anti-CD33 ADC with dose-limiting neutropenia in cynomolgus monkeys. We tested our model by comparing PK predictions based on the unconjugated antibody to observed ADC PK data that was not utilized for model development. Prospective prediction of human PK was performed by incorporating in vitro binding affinity differences between species for varying levels of CD33 target expression. Additionally, this approach was used to predict human PK of other previously tested anti-CD33 molecules with published clinical data. The findings showed that, for a cytotoxic ADC with non-linear PK and limited preclinical PK data, incorporating RO in the PK model and using data from the corresponding unconjugated antibody at higher doses allowed the identification of parameters to characterize monkey PK and enabled human PK predictions.
Collapse
Affiliation(s)
| | - Doug Leipold
- Preclinical Translational Pharmacokinetics Department
| | | | | | | | | | | | | | - Melissa Schutten
- Safety Assessment Department Genentech Inc., South San Francisco, CA, USA
| | - Hong Wang
- Safety Assessment Department Genentech Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
66
|
Gandhi AV, Arlotta KJ, Chen HN, Owen SC, Carpenter JF. Biophysical Properties and Heating-Induced Aggregation of Lysine-Conjugated Antibody-Drug Conjugates. J Pharm Sci 2018; 107:1858-1869. [DOI: 10.1016/j.xphs.2018.03.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/17/2018] [Accepted: 03/27/2018] [Indexed: 12/12/2022]
|
67
|
Tsumura R, Manabe S, Takashima H, Koga Y, Yasunaga M, Matsumura Y. Influence of the dissociation rate constant on the intra-tumor distribution of antibody-drug conjugate against tissue factor. J Control Release 2018; 284:49-56. [PMID: 29906553 DOI: 10.1016/j.jconrel.2018.06.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/07/2018] [Accepted: 06/11/2018] [Indexed: 10/14/2022]
Abstract
Antibody-drug conjugates (ADCs) are currently considered to be promising agents for cancer therapy. However, especially in solid tumors, the uneven distribution of ADCs would decrease their efficacy in clinical studies. We suggest that in addition to optimizing ADC components, such as the linker structure and anticancer agent, it is necessary to consider the distribution of the ADC within tumor tissue. In this study, we established three kinds of anti-tissue factor (TF) ADCs: 1849ADC with a low kd, 444ADC with an intermediate kd, and 1084ADC with a high kd. All three of the anti-TF ADCs exhibited almost the same in vitro cytotoxicity and pharmacological and biochemical characteristics, although the binding kinetics parameters differed. In vivo, all ADCs exerted equivalent antitumor effects against small BxPC3 tumors. However, on larger BxPC3 tumors, 1084ADC (higher kd) exerted higher antitumor activity than 1849ADC (lower kd). Furthermore, immunofluorescence staining indicated that 1084ADC was distributed throughout the whole tumor, whereas 1849ADC was mainly localized close to tumor vessels. We conclude that the ADC with a higher kd increased the antitumor effect of because it penetrated and distributed evenly throughout the entire solid tumor. These findings highlight the importance of the kd of a mAb in ADC design.
Collapse
Affiliation(s)
- Ryo Tsumura
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa, Japan.
| | - Shino Manabe
- Synthetic Cellular Chemistry Laboratory, RIKEN, Wako, Japan.
| | - Hiroki Takashima
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa, Japan.
| | - Yoshikatsu Koga
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa, Japan.
| | - Masahiro Yasunaga
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa, Japan.
| | - Yasuhiro Matsumura
- Division of Developmental Therapeutics, EPOC, National Cancer Center, Kashiwa, Japan.
| |
Collapse
|
68
|
Shao S, Tsai MH, Lu J, Yu T, Jin J, Xiao D, Jiang H, Han M, Wang M, Wang J. Site-specific and hydrophilic ADCs through disulfide-bridged linker and branched PEG. Bioorg Med Chem Lett 2018; 28:1363-1370. [DOI: 10.1016/j.bmcl.2018.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 02/28/2018] [Accepted: 03/02/2018] [Indexed: 01/19/2023]
|
69
|
Warnders FJ, Lub-de Hooge MN, de Vries EGE, Kosterink JGW. Influence of protein properties and protein modification on biodistribution and tumor uptake of anticancer antibodies, antibody derivatives, and non-Ig scaffolds. Med Res Rev 2018; 38:1837-1873. [PMID: 29635825 DOI: 10.1002/med.21498] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/30/2018] [Accepted: 03/02/2018] [Indexed: 12/11/2022]
Abstract
Newly developed protein drugs that target tumor-associated antigens are often modified in order to increase their therapeutic effect, tumor exposure, and safety profile. During the development of protein drugs, molecular imaging is increasingly used to provide additional information on their in vivo behavior. As a result, there are increasing numbers of studies that demonstrate the effect of protein modification on whole body distribution and tumor uptake of protein drugs. However, much still remains unclear about how to interpret obtained biodistribution data correctly. Consequently, there is a need for more insight in the correct way of interpreting preclinical and clinical imaging data. Summarizing the knowledge gained to date may facilitate this interpretation. This review therefore provides an overview of specific protein properties and modifications that can affect biodistribution and tumor uptake of anticancer antibodies, antibody fragments, and nonimmunoglobulin scaffolds. Protein properties that are discussed in this review are molecular size, target interaction, FcRn binding, and charge. Protein modifications that are discussed are radiolabeling, fluorescent labeling drug conjugation, glycosylation, humanization, albumin binding, and polyethylene glycolation.
Collapse
Affiliation(s)
- Frank-Jan Warnders
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jos G W Kosterink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,PharmacoTherapy, Epidemiology & Economy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
70
|
Zettlitz KA, Tsai WTK, Knowles SM, Kobayashi N, Donahue TR, Reiter RE, Wu AM. Dual-Modality Immuno-PET and Near-Infrared Fluorescence Imaging of Pancreatic Cancer Using an Anti-Prostate Stem Cell Antigen Cys-Diabody. J Nucl Med 2018; 59:1398-1405. [PMID: 29602820 DOI: 10.2967/jnumed.117.207332] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/12/2018] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer has a high mortality rate due to late diagnosis and the tendency to invade surrounding tissues and metastasize at an early stage. A molecular imaging agent that enables both presurgery antigen-specific PET (immuno-PET) and intraoperative near-infrared fluorescence (NIRF) guidance might benefit diagnosis of pancreatic cancer, staging, and surgical resection, which remains the only curative treatment. Methods: We developed a dual-labeled probe based on A2 cys-diabody (A2cDb) targeting the cell-surface prostate stem cell antigen (PSCA), which is expressed in most pancreatic cancers. Maleimide-IRDye800CW was site-specifically conjugated to the C-terminal cys-tag (A2cDb-800) without impairing integrity or affinity (half-maximal binding, 4.3 nM). Direct radioiodination with 124I (124I-A2cDb-800) yielded a specific activity of 159 ± 48 MBq/mg with a radiochemical purity exceeding 99% and 65% ± 4.5% immunoreactivity (n = 3). In vivo specificity for PSCA-expressing tumor cells and biodistribution of the dual-modality tracer were evaluated in a prostate cancer xenograft model and compared with single-labeled 124I-A2cDb. Patient-derived pancreatic ductal adenocarcinoma xenografts (PDX-PDACs) were grown subcutaneously in NSG mice and screened for PSCA expression by immuno-PET. Small-animal PET/CT scans of PDX-PDAC-bearing mice were obtained using the dual-modality 124I-A2cDb-800 followed by postmortem NIRF imaging with the skin removed. Tumors and organs were analyzed ex vivo to compare the relative fluorescent signals without obstruction by other organs. Results: Specific uptake in PSCA-positive tumors and low nonspecific background activity resulted in high-contrast immuno-PET images. Concurrent with the PET studies, fluorescent signal was observed in the PSCA-positive tumors of mice injected with the dual-tracer 124I-A2cDb-800, with low background uptake or autofluorescence in the surrounding tissue. Ex vivo biodistribution confirmed comparable tumor uptake of both 124I-A2cDb-800 and 124I-A2cDb. Conclusion: Dual-modality imaging using the anti-PSCA cys-diabody resulted in high-contrast immuno-PET/NIRF images of PDX-PDACs, suggesting that this imaging agent might offer both noninvasive whole-body imaging to localize PSCA-positive pancreatic cancer and fluorescence image-guided identification of tumor margins during surgery.
Collapse
Affiliation(s)
- Kirstin A Zettlitz
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California .,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Wen-Ting K Tsai
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Scott M Knowles
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Naoko Kobayashi
- David Geffen School of Medicine, UCLA, Los Angeles, California.,Department of Urology, UCLA, Los Angeles, California; and
| | - Timothy R Donahue
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California.,Division of General Surgery, Department of Surgery, UCLA, Los Angeles, California
| | - Robert E Reiter
- David Geffen School of Medicine, UCLA, Los Angeles, California.,Department of Urology, UCLA, Los Angeles, California; and
| | - Anna M Wu
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, California.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California.,David Geffen School of Medicine, UCLA, Los Angeles, California
| |
Collapse
|
71
|
A Polar Sulfamide Spacer Significantly Enhances the Manufacturability, Stability, and Therapeutic Index of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:antib7010012. [PMID: 31544864 PMCID: PMC6698870 DOI: 10.3390/antib7010012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/08/2018] [Accepted: 02/17/2018] [Indexed: 11/17/2022] Open
Abstract
Despite tremendous efforts in the field of targeted cancer therapy with antibody–drug conjugates (ADCs), attrition rates have been high. Historically, the priority in ADC development has been the selection of target, antibody, and toxin, with little focus on the nature of the linker. We show here that a short and polar sulfamide spacer (HydraSpace™, Oss, The Netherlands) positively impacts ADC properties in various ways: (a) efficiency of conjugation; (b) stability; and (c) therapeutic index. Different ADC formats are explored in terms of drug-to-antibody ratios (DAR2, DAR4) and we describe the generation of a DAR4 ADC by site-specific attachment of a bivalent linker–payload construct to a single conjugation site in the antibody. A head-to-head comparison of HydraSpace™-containing DAR4 ADCs to marketed drugs, derived from the same antibody and toxic payload components, indicated a significant improvement in both the efficacy and safety of several vivo models, corroborated by in-depth pharmacokinetic analysis. Taken together, HydraSpace™ technology based on a polar sulfamide spacer provides significant improvement in manufacturability, stability, and ADC design, and is a powerful platform to enable next-generation ADCs with enhanced therapeutic index.
Collapse
|
72
|
Lucas AT, Price LSL, Schorzman AN, Storrie M, Piscitelli JA, Razo J, Zamboni WC. Factors Affecting the Pharmacology of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:E10. [PMID: 31544862 PMCID: PMC6698819 DOI: 10.3390/antib7010010] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Major advances in therapeutic proteins, including antibody-drug conjugates (ADCs), have created revolutionary drug delivery systems in cancer over the past decade. While these immunoconjugate agents provide several advantages compared to their small-molecule counterparts, their clinical use is still in its infancy. The considerations in their development and clinical use are complex, and consist of multiple components and variables that can affect the pharmacologic characteristics. It is critical to understand the mechanisms employed by ADCs in navigating biological barriers and how these factors affect their biodistribution, delivery to tumors, efficacy, and toxicity. Thus, future studies are warranted to better understand the complex pharmacology and interaction between ADC carriers and biological systems, such as the mononuclear phagocyte system (MPS) and tumor microenvironment. This review provides an overview of factors that affect the pharmacologic profiles of ADC therapies that are currently in clinical use and development.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Lauren S L Price
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Mallory Storrie
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | | | - Juan Razo
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
73
|
Abstract
Site-specific conjugation methods are becoming increasingly important in building next-generation antibody-drug conjugates. We have developed a site-specific conjugation technology based on monoclonal antibodies with engineered selenocysteine (Sec) residues, named selenomabs. Here, we provide protocols for the engineering, expression, and purification of selenomabs in single-chain variable fragment (scFv)-Fc format. Methods for selective conjugation of selenomabs to selenol-reactive compounds and analytical characterization of selenomab conjugates are also included.
Collapse
|
74
|
Wagh A, Song H, Zeng M, Tao L, Das TK. Challenges and new frontiers in analytical characterization of antibody-drug conjugates. MAbs 2018; 10:222-243. [PMID: 29293399 DOI: 10.1080/19420862.2017.1412025] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a growing class of biotherapeutics in which a potent small molecule is linked to an antibody. ADCs are highly complex and structurally heterogeneous, typically containing numerous product-related species. One of the most impactful steps in ADC development is the identification of critical quality attributes to determine product characteristics that may affect safety and efficacy. However, due to the additional complexity of ADCs relative to the parent antibodies, establishing a solid understanding of the major quality attributes and determining their criticality are a major undertaking in ADC development. Here, we review the development challenges, especially for reliable detection of quality attributes, citing literature and new data from our laboratories, highlight recent improvements in major analytical techniques for ADC characterization and control, and discuss newer techniques, such as two-dimensional liquid chromatography, that have potential to be included in analytical control strategies.
Collapse
Affiliation(s)
- Anil Wagh
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Hangtian Song
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Ming Zeng
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Li Tao
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| | - Tapan K Das
- a Molecular & Analytical Development , Bristol-Myers Squibb , New Jersey , USA
| |
Collapse
|
75
|
Gupta S, Drolet DW, Wolk SK, Waugh SM, Rohloff JC, Carter JD, Mayfield WS, Otis MR, Fowler CR, Suzuki T, Hirota M, Ishikawa Y, Schneider DJ, Janjic N. Pharmacokinetic Properties of DNA Aptamers with Base Modifications. Nucleic Acid Ther 2017; 27:345-353. [PMID: 28961063 PMCID: PMC5706628 DOI: 10.1089/nat.2017.0683] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The addition of novel side chains at the 5-position of uracil is an effective means to increase chemical diversity of aptamers and hence the success rate for discovery of high-affinity ligands to protein targets. Such modifications also increase nuclease resistance, which is useful in a range of applications, especially for therapeutics. In this study, we assess the impact of these side chains on plasma pharmacokinetics of modified aptamers conjugated to a 40 kDa polyethylene glycol. We show that clearance from plasma depends on relative hydrophobicity: side chains with a negative cLogP (more hydrophilic) result in slower plasma clearance compared with side chains with a positive cLogP (more hydrophobic). We show that clearance increases with the number of side chains in sequences of ≥28 synthons, but this effect is dramatically diminished in shorter sequences. These results serve as a guide for the design of new therapeutic aptamers with diversity-enhancing side chains.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tomoki Suzuki
- 2 Otsuka Pharmaceutical Co., Ltd. , Tokushima, Japan
| | - Masao Hirota
- 2 Otsuka Pharmaceutical Co., Ltd. , Tokushima, Japan
| | | | | | | |
Collapse
|
76
|
Malik P, Phipps C, Edginton A, Blay J. Pharmacokinetic Considerations for Antibody-Drug Conjugates against Cancer. Pharm Res 2017; 34:2579-2595. [PMID: 28924691 DOI: 10.1007/s11095-017-2259-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/09/2017] [Indexed: 12/26/2022]
Abstract
Antibody-drug conjugates (ADCs) are ushering in the next era of targeted therapy against cancer. An ADC for cancer therapy consists of a potent cytotoxic payload that is attached to a tumour-targeted antibody by a chemical linker, usually with an average drug-to-antibody ratio (DAR) of 3.5-4. The theory is to deliver potent cytotoxic payloads directly to tumour cells while sparing healthy cells. However, practical application has proven to be more difficult. At present there are only two ADCs approved for clinical use. Nevertheless, in the last decade there has been an explosion of options for ADC engineering to optimize target selection, Fc receptor interactions, linker, payload and more. Evaluation of these strategies requires an understanding of the mechanistic underpinnings of ADC pharmacokinetics. Development of ADCs for use in cancer further requires an understanding of tumour properties and kinetics within the tumour environment, and how the presence of cancer as a disease will impact distribution and elimination. Key pharmacokinetic considerations for the successful design and clinical application of ADCs in oncology are explored in this review, with a focus on the mechanistic determinants of distribution and elimination.
Collapse
Affiliation(s)
- Paul Malik
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada
| | - Colin Phipps
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada.,DMPK & Translational Modeling, Abbvie Inc., North Chicago, Illinois, 60064, USA
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada.
| | - Jonathan Blay
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada
| |
Collapse
|
77
|
Durbin KR, Nottoli MS, Catron ND, Richwine N, Jenkins GJ. High-Throughput, Multispecies, Parallelized Plasma Stability Assay for the Determination and Characterization of Antibody-Drug Conjugate Aggregation and Drug Release. ACS OMEGA 2017; 2:4207-4215. [PMID: 30023717 PMCID: PMC6044903 DOI: 10.1021/acsomega.7b00452] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/20/2017] [Indexed: 06/08/2023]
Abstract
The stability of antibody-drug conjugates (ADCs) in circulation is critical for maximum efficacy and minimal toxicity. An ADC reaching the intended target intact can deliver the highest possible drug load to the tumor and reduce off-target toxicity from free drug in the blood. As such, assessment of ADC stability is a vital piece of data during development. However, traditional ADC stability assays can be manually intensive, low-throughput, and require large quantities of ADC material. Here, we introduce an automated, high-throughput plasma stability assay for screening drug release and aggregation over 144 h for up to 40 ADCs across five matrices simultaneously. The amount of ADC material during early drug development is often limited, so this assay was implemented in 384-well format to minimize material requirements to <100 μg of each ADC and 100 μL of plasma per species type. Drug release and aggregation output were modeled using nonlinear regression equations to calculate formation rates for each data type. A set of 15 ADCs with different antibodies and identical valine-citrulline-p-aminobenzylcarbamate-monomethylauristatin E linker-drug payloads was tested and formation rates were compared across ADCs and between species, revealing several noteworthy trends. In particular, a wide range in aggregation was found when altering only the antibody, suggesting a key role for plasma stability screening early in the development process to find and remove antibody candidates with the potential to create unstable ADCs. The assay presented here can be leveraged to provide stability data on new chemistry and antibody screening initiatives, select the best candidate for in vivo studies, and provide results that highlight stability issues inherent to particular ADC designs throughout all stages of ADC development.
Collapse
Affiliation(s)
- Kenneth R. Durbin
- Drug
Metabolism and Pharmacokinetics and Drug Product Development, AbbVie, Inc., 1 N. Waukegan
Drive, North Chicago, Illinois 60064, United
States
| | - M. Shannon Nottoli
- Drug
Metabolism and Pharmacokinetics and Drug Product Development, AbbVie, Inc., 1 N. Waukegan
Drive, North Chicago, Illinois 60064, United
States
| | - Nathaniel D. Catron
- Drug
Metabolism and Pharmacokinetics and Drug Product Development, AbbVie, Inc., 1 N. Waukegan
Drive, North Chicago, Illinois 60064, United
States
| | - Nicole Richwine
- Drug
Metabolism and Pharmacokinetics and Drug Product Development, AbbVie, Inc., 1 N. Waukegan
Drive, North Chicago, Illinois 60064, United
States
| | - Gary J. Jenkins
- Drug
Metabolism and Pharmacokinetics and Drug Product Development, AbbVie, Inc., 1 N. Waukegan
Drive, North Chicago, Illinois 60064, United
States
| |
Collapse
|
78
|
Jacobson O, Li Q, Chen H, Niu G, Kiesewetter DO, Xu L, Cook K, Yang G, Dall'Acqua W, Tsui P, Peng L, Chen X. PET-Guided Evaluation and Optimization of Internalized Antibody-Drug Conjugates Targeting Erythropoietin-Producing Hepatoma A2 Receptor. J Nucl Med 2017; 58:1838-1844. [PMID: 28546337 DOI: 10.2967/jnumed.117.192245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/04/2017] [Indexed: 12/26/2022] Open
Abstract
The erythropoietin-producing hepatoma A2 receptor (EphA2) is a tyrosine kinase overexpressed by tumor stroma and cancer cells. A high expression level of EphA2 predicts poor prognosis, correlating with disease progression and metastasis. Therefore, EphA2 is a relevant therapeutic target for human cancer. Antibodies, selectively bound to EphA2, can induce rapid receptor phosphorylation that results in antibody internalization and degradation. This internalization mechanism has been exploited with the development of antibody-drug conjugates (ADCs) for cancer chemotherapy. In this study, we used PET imaging to study the pharmacokinetics and tumor delivery of a panel of anti-EphA2 monoclonal antibodies (mAbs) with and without drug conjugates. Methods: A library of human anti-EphA2 mAbs were screened and evaluated for EphA2 internalization rate, binding affinity, epitope binding, and hydrophobicity. We chose 3 of these antibodies, denoted as 1C1, 3B10, and 2H7, which recognize different epitopes, for further evaluation. ADCs were generated by S239C mutation to give a ratio of 2 drug molecules per antibody. Native mAbs and ADCs were characterized, after conjugation to a DFO chelator and 89Zr radiolabeling, in assays including cell uptake, internalization, hydrophobicity, and in vivo imaging using PET. Results: All 3 mAbs had high affinities for EphA2 but exhibited different internalization rates following the order of 1C1 > 3B10 > 2H7. Internalization rate is only 1 factor that affects in vitro cell uptake and in vivo tumor accumulation. Interestingly, the hydrophobicity of the mAbs, which followed the order of 2H7 > 1C1 > 3B10, had a strong correlation with in vivo tumor uptake measured by PET, with the least hydrophobic antibody, 3B10, showing the highest tumor uptake. ADC significantly reduced the in vivo uptake of all 3 mAbs. Conclusion: Tumor uptake of mAb is a complex process that is affected by multiple parameters, including internalization, hydrophobicity, and chemical modification. Our results suggest that the addition of drug molecules to mAb increases the clearance of the mAb presumably due to the increased hydrophobicity. Understanding the complexity of antibody-based tumor delivery may help improve ADC engineering for better tumor targeting and reduced side effects.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Qing Li
- Department of Antibody Discovery & Protein Engineering, MedImmune LLC, Gaithersburg, Maryland
| | - Haojun Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Dale O Kiesewetter
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| | - Lan Xu
- Department of Antibody Discovery & Protein Engineering, MedImmune LLC, Gaithersburg, Maryland
| | - Kimberly Cook
- Department of Antibody Discovery & Protein Engineering, MedImmune LLC, Gaithersburg, Maryland
| | - Gengcheng Yang
- Department of Antibody Discovery & Protein Engineering, MedImmune LLC, Gaithersburg, Maryland
| | - William Dall'Acqua
- Department of Antibody Discovery & Protein Engineering, MedImmune LLC, Gaithersburg, Maryland
| | - Ping Tsui
- Department of Antibody Discovery & Protein Engineering, MedImmune LLC, Gaithersburg, Maryland
| | - Li Peng
- Department of Antibody Discovery & Protein Engineering, MedImmune LLC, Gaithersburg, Maryland
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
79
|
Cilliers C, Nessler I, Christodolu N, Thurber GM. Tracking Antibody Distribution with Near-Infrared Fluorescent Dyes: Impact of Dye Structure and Degree of Labeling on Plasma Clearance. Mol Pharm 2017; 14:1623-1633. [PMID: 28294622 PMCID: PMC5415873 DOI: 10.1021/acs.molpharmaceut.6b01091] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Monoclonal
antibodies labeled with near-infrared (NIR) fluorophores
have potential use in disease detection, intraoperative imaging, and
pharmacokinetic characterization of therapeutic antibodies in both
the preclinical and clinical setting. Recent work has shown conjugation
of NIR fluorophores to antibodies can potentially alter antibody disposition
at a sufficiently high degree of labeling (DoL); however, other reports
show minimal impact after labeling with NIR fluorophores. In this
work, we label two clinically approved antibodies, Herceptin (trastuzumab)
and Avastin (bevacizumab), with NIR dyes IRDye 800CW (800CW) or Alexa
Fluor 680 (AF680), at 1.2 and 0.3 dyes/antibody and examine the impact
of fluorophore conjugation on antibody plasma clearance and tissue
distribution. At 0.3 DoL, AF680 conjugates exhibited similar clearance
to unlabeled antibody over 17 days while 800CW conjugates diverged
after 4 days, suggesting AF680 is a more suitable choice for long-term
pharmacokinetic studies. At the 1.2 DoL, 800CW conjugates cleared
faster than unlabeled antibodies after several hours, in agreement
with other published reports. The tissue biodistribution for bevacizumab–800CW
and −AF680 conjugates agreed well with literature reported
biodistributions using radiolabels. However, the greater tissue autofluorescence
at 680 nm resulted in limited detection above background at low (∼2
mg/kg) doses and 0.3 DoL for AF680, indicating that 800CW is more
appropriate for short-term biodistribution measurements and intraoperative
imaging. Overall, our work shows a DoL of 0.3 or less for non-site-specifically
labeled antibodies (with a Poisson distribution) is ideal for limiting
the impact of NIR fluorophores on antibody pharmacokinetics.
Collapse
Affiliation(s)
- Cornelius Cilliers
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Ian Nessler
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Nikolas Christodolu
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Greg M Thurber
- Department of Chemical Engineering and ‡Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| |
Collapse
|
80
|
Newman DJ, Cragg GM. Current Status of Marine-Derived Compounds as Warheads in Anti-Tumor Drug Candidates. Mar Drugs 2017; 15:md15040099. [PMID: 28353637 PMCID: PMC5408245 DOI: 10.3390/md15040099] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 01/09/2023] Open
Abstract
In this review, we have attempted to describe all of the antibody–drug conjugates using a marine-derived compound as the “warhead”, that are currently in clinical trials as listed in the current version of the NIH clinical trials database (clinicaltrials.gov). In searching this database, we used the beta-test version currently available, as it permitted more specific search parameters, since the regular version did not always find trials that had been completed in the past with some agents. We also added small discussion sections on candidates that are still at the preclinical stage, including a derivative of diazonamide that has an unusual interaction with tubulin (DZ-23840), which may also be a potential warhead in the future.
Collapse
|
81
|
Li X, Nelson CG, Nair RR, Hazlehurst L, Moroni T, Martinez-Acedo P, Nanna AR, Hymel D, Burke TR, Rader C. Stable and Potent Selenomab-Drug Conjugates. Cell Chem Biol 2017; 24:433-442.e6. [PMID: 28330604 DOI: 10.1016/j.chembiol.2017.02.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 12/16/2016] [Accepted: 02/10/2017] [Indexed: 02/06/2023]
Abstract
Selenomabs are engineered monoclonal antibodies with one or more translationally incorporated selenocysteine residues. The unique reactivity of the selenol group of selenocysteine permits site-specific conjugation of drugs. Compared with other natural and unnatural amino acid and carbohydrate residues that have been used for the generation of site-specific antibody-drug conjugates, selenocysteine is particularly reactive, permitting fast, single-step, and efficient reactions under near physiological conditions. Using a tailored conjugation chemistry, we generated highly stable selenomab-drug conjugates and demonstrated their potency and selectivity in vitro and in vivo. These site-specific antibody-drug conjugates built on a selenocysteine interface revealed broad therapeutic utility in liquid and solid malignancy models.
Collapse
Affiliation(s)
- Xiuling Li
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Christopher G Nelson
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Rajesh R Nair
- Molecular Oncology Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Lori Hazlehurst
- Molecular Oncology Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Tina Moroni
- Proteomics and Mass Spectrometry Core, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Pablo Martinez-Acedo
- Proteomics and Mass Spectrometry Core, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Alex R Nanna
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - David Hymel
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Terrence R Burke
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Christoph Rader
- Department of Cancer Biology, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
82
|
Nunes JPM, Vassileva V, Robinson E, Morais M, Smith MEB, Pedley RB, Caddick S, Baker JR, Chudasama V. Use of a next generation maleimide in combination with THIOMAB™ antibody technology delivers a highly stable, potent and near homogeneous THIOMAB™ antibody-drug conjugate (TDC). RSC Adv 2017. [DOI: 10.1039/c7ra04606e] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Conjugation of next generation maleimides to engineered cysteines in a THIOMAB™ antibody delivers a highly stable and potent THIOMAB™ antibody-drug conjugate.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - James R. Baker
- Department of Chemistry
- University College London
- London
- UK
| | - Vijay Chudasama
- Department of Chemistry
- University College London
- London
- UK
- Research Institute for Medicines (iMed.ULisboa)
| |
Collapse
|
83
|
Robinson E, Nunes JPM, Vassileva V, Maruani A, Nogueira JCF, Smith MEB, Pedley RB, Caddick S, Baker JR, Chudasama V. Pyridazinediones deliver potent, stable, targeted and efficacious antibody–drug conjugates (ADCs) with a controlled loading of 4 drugs per antibody. RSC Adv 2017. [DOI: 10.1039/c7ra00788d] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Delivering potent, stable, targeted and in vivo efficacious antibody–drug conjugates (ADCs) using pyridazinedione functional disulfide re-bridging reagents.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - James R. Baker
- Department of Chemistry
- University College London
- London
- UK
| | | |
Collapse
|
84
|
Morais M, Nunes JPM, Karu K, Forte N, Benni I, Smith MEB, Caddick S, Chudasama V, Baker JR. Optimisation of the dibromomaleimide (DBM) platform for native antibody conjugation by accelerated post-conjugation hydrolysis. Org Biomol Chem 2017; 15:2947-2952. [DOI: 10.1039/c7ob00220c] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dibromomaleimide (DBM) reagents are described which hydrolyse rapidly post-conjugation, representing an optimised platform for homogeneous and stable antibody conjugation.
Collapse
|
85
|
Adumeau P, Sharma SK, Brent C, Zeglis BM. Site-Specifically Labeled Immunoconjugates for Molecular Imaging--Part 1: Cysteine Residues and Glycans. Mol Imaging Biol 2016; 18:1-17. [PMID: 26754790 PMCID: PMC4722084 DOI: 10.1007/s11307-015-0919-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Due to their remarkable selectivity and specificity for cancer biomarkers, immunoconjugates have emerged as extremely promising vectors for the delivery of diagnostic radioisotopes and fluorophores to malignant tissues. Paradoxically, however, these tools for precision medicine are synthesized in a remarkably imprecise way. Indeed, the vast majority of immunoconjugates are created via the random conjugation of bifunctional probes (e.g., DOTA-NCS) to amino acids within the antibody (e.g., lysines). Yet antibodies have multiple copies of these residues throughout their macromolecular structure, making control over the location of the conjugation reaction impossible. This lack of site specificity can lead to the formation of poorly defined, heterogeneous immunoconjugates with suboptimal in vivo behavior. Over the past decade, interest in the synthesis and development of site-specifically labeled immunoconjugates—both antibody-drug conjugates as well as constructs for in vivo imaging—has increased dramatically, and a number of reports have suggested that these better defined, more homogeneous constructs exhibit improved performance in vivo compared to their randomly modified cousins. In this two-part review, we seek to provide an overview of the various methods that have been developed to create site-specifically modified immunoconjugates for positron emission tomography, single photon emission computed tomography, and fluorescence imaging. We will begin with an introduction to the structure of antibodies and antibody fragments. This is followed by the core of the work: sections detailing the four different approaches to site-specific modification strategies based on cysteine residues, glycans, peptide tags, and unnatural amino acids. These discussions will be divided into two installments: cysteine residues and glycans will be detailed in Part 1 of the review, while peptide tags and unnatural amino acids will be addressed in Part 2. Ultimately, we sincerely hope that this review fosters interest and enthusiasm for site-specific immunoconjugates within the nuclear medicine and molecular imaging communities.
Collapse
Affiliation(s)
- Pierre Adumeau
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Sai Kiran Sharma
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY10065, NY, USA
| | - Colleen Brent
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Brian M Zeglis
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA.
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY10065, NY, USA.
| |
Collapse
|
86
|
Sukumaran S, Zhang C, Leipold DD, Saad OM, Xu K, Gadkar K, Samineni D, Wang B, Milojic-Blair M, Carrasco-Triguero M, Rubinfeld B, Fielder P, Lin K, Ramanujan S. Development and Translational Application of an Integrated, Mechanistic Model of Antibody-Drug Conjugate Pharmacokinetics. AAPS JOURNAL 2016; 19:130-140. [PMID: 27679517 DOI: 10.1208/s12248-016-9993-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/13/2016] [Indexed: 01/07/2023]
Abstract
Antibody drug conjugates (ADC), in which small molecule cytotoxic agents are non-specifically linked to antibodies, can enable targeted delivery of chemotherapeutics to tumor cells. ADCs are often produced and administered as a mixture of conjugated antibodies with different drug to antibody ratios (DAR) resulting in complex and heterogeneous disposition kinetics. We developed a mechanism-based platform model that can describe and predict the complex pharmacokinetic (PK) behavior of ADCs with protease-cleavable valine-citrulline (VC) linker linked to Monomethylmonomethyl auristatin F/E by incorporating known mechanisms of ADC disposition. The model includes explicit representation of all DAR species; DAR-dependent sequential deconjugation of the drug, resulting in the conversion of higher DAR to lower DAR species; and DAR-dependent antibody/ADC clearance. PK profiles of multiple analytes (total antibody, drug-conjugated antibody, and/or antibody-conjugated drug) for different ADC molecules and targets in rodents and cynomolgus monkeys were used for model development. The integrated cross-species model was successful in capturing the multi-analyte PK profiles after administration of purified ADCs with defined DAR species and ADCs with mixtures of DAR. Human PK predictions for DSTP3086S (anti-STEAP1-vc-MMAE) with the platform model agreed well with PK (total antibody and antibody-conjugated drug concentrations) measurements in the dose-ranging phase I clinical study. The integrated model is applicable to various other ADCs with different formats, conjugated drugs, and linkers, and provides a valuable tool for the exploration of mechanisms governing disposition of ADCs and enables translational predictions.
Collapse
Affiliation(s)
- Siddharth Sukumaran
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Crystal Zhang
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Douglas D Leipold
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Ola M Saad
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Keyang Xu
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Kapil Gadkar
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Divya Samineni
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Bei Wang
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Marija Milojic-Blair
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | | | - Bonnee Rubinfeld
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Paul Fielder
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Kedan Lin
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Saroja Ramanujan
- Genentech Research and Early Development, 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
87
|
Patterson JT, Wilson HD, Asano S, Nilchan N, Fuller RP, Roush WR, Rader C, Barbas CF. Human Serum Albumin Domain I Fusion Protein for Antibody Conjugation. Bioconjug Chem 2016; 27:2271-2275. [PMID: 27666414 DOI: 10.1021/acs.bioconjchem.6b00432] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bioorthogonal labeling of antibodies enables the conjugation of compounds, such as small molecules or peptides, which expand targeting capacity or enhance cytotoxicity. Taking advantage of a cyclohexene sulfonamide compound that site-selectively labels Lys64 in human serum albumin (HSA), we demonstrate that domain I of HSA can be used as a fusion protein for the preparation of antibody conjugates. Trastuzumab fusions were expressed at the N-terminus of the light chain or the C-terminus of the heavy chain enabling conjugation to small molecules. Moreover, these conjugates retained HER2 binding and proved to be highly stable in human plasma. Antibody conjugation via HSA domain I fusion should therefore have broad utility for making serum-stable antibody conjugates, particularly for antibody-drug conjugates.
Collapse
Affiliation(s)
- James T Patterson
- Department of Chemistry, Department of Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Henry D Wilson
- Department of Cancer Biology, Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida , Jupiter, Florida 33458, United States
| | - Shigehiro Asano
- Department of Chemistry, Department of Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Napon Nilchan
- Department of Chemistry, The Scripps Research Institute, Scripps Florida , Jupiter, Florida 33458, United States
| | - Roberta P Fuller
- Department of Chemistry, Department of Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| | - William R Roush
- Department of Chemistry, The Scripps Research Institute, Scripps Florida , Jupiter, Florida 33458, United States
| | - Christoph Rader
- Department of Cancer Biology, Department of Molecular Therapeutics, The Scripps Research Institute, Scripps Florida , Jupiter, Florida 33458, United States
| | - Carlos F Barbas
- Department of Chemistry, Department of Cell and Molecular Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| |
Collapse
|
88
|
Strop P, Tran TT, Dorywalska M, Delaria K, Dushin R, Wong OK, Ho WH, Zhou D, Wu A, Kraynov E, Aschenbrenner L, Han B, O'Donnell CJ, Pons J, Rajpal A, Shelton DL, Liu SH. RN927C, a Site-Specific Trop-2 Antibody-Drug Conjugate (ADC) with Enhanced Stability, Is Highly Efficacious in Preclinical Solid Tumor Models. Mol Cancer Ther 2016; 15:2698-2708. [PMID: 27582525 DOI: 10.1158/1535-7163.mct-16-0431] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/12/2016] [Indexed: 11/16/2022]
Abstract
Trop-2, also known as TACSTD2, EGP-1, GA733-1, and M1S1, is frequently expressed on a variety of human carcinomas, and its expression is often associated with poor prognosis of the diseases. However, it is also present on the epithelium of several normal tissues. A comprehensively designed Trop-2-targeting antibody-drug conjugate (ADC), balancing both efficacy and toxicity, is therefore necessary to achieve clinical utility. To this end, we developed a cleavable Trop-2 ADC (RN927C) using a site-specific transglutaminase-mediated conjugation method and a proprietary microtubule inhibitor (MTI) linker-payload, PF-06380101. Robust in vitro cytotoxicity of RN927C was observed on a panel of Trop-2-expressing tumor cell lines, with IC50 generally in the subnanomolar range. As expected for an MTI-containing ADC, RN927C readily induced mitotic arrest of treated cells in vitro and in vivo, followed by subsequent cell death. The in vivo efficacy of RN927C was tested in multiple cell line and patient-derived xenograft tumor models, including pancreatic, lung, ovarian, and triple-negative breast tumor types. Single-dose administration of RN927C at 0.75 to 3 mg/kg was generally sufficient to induce sustained regression of Trop-2-expressing tumors and showed superior efficacy over standard treatment with paclitaxel or gemcitabine. Administration of RN927C in nonhuman primate toxicity studies resulted in target-mediated effects in skin and oral mucosa, consistent with Trop-2 expression in these epithelial tissues with minimal, non-dose limiting off-target toxicities. On the basis of the combined efficacy and safety results, RN927C is postulated to have a favorable therapeutic index for treatment of solid tumors. Mol Cancer Ther; 15(11); 2698-708. ©2016 AACR.
Collapse
Affiliation(s)
- Pavel Strop
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | - Thomas-Toan Tran
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | | | - Kathy Delaria
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | - Russell Dushin
- Worldwide Medicinal Chemistry, Pfizer Inc., Groton, Connecticut
| | - Oi Kwan Wong
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | - Wei-Hsien Ho
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | - Dahui Zhou
- Worldwide Medicinal Chemistry, Pfizer Inc., Groton, Connecticut
| | - Aidong Wu
- Pharmacokinetics, Dynamics & Metabolism, Pfizer Inc., San Diego, California
| | - Eugenia Kraynov
- Pharmacokinetics, Dynamics & Metabolism, Pfizer Inc., San Diego, California
| | | | - Bora Han
- Drug Safety R&D, Pfizer Inc., San Diego, California
| | | | - Jaume Pons
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | - Arvind Rajpal
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | - Dave L Shelton
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California
| | - Shu-Hui Liu
- Oncology-Rinat R&D, Pfizer Inc., South San Francisco, California.
| |
Collapse
|
89
|
Liu B, Guo H, Zhang J, Xue J, Yang Y, Qin T, Xu J, Guo Q, Zhang D, Qian W, Li B, Hou S, Dai J, Guo Y, Wang H. In-Depth Characterization of a Pro-Antibody-Drug Conjugate by LC-MS. Mol Pharm 2016; 13:2702-10. [PMID: 27377124 DOI: 10.1021/acs.molpharmaceut.6b00280] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pro-antibody-drug conjugate (PDC) is a hybrid structural format of immunoconjugate, where the structural complexity of pro-antibody and intrinsic heterogeneity of ADCs impose a prominent analytical challenge to the in-depth characterization of PDCs. In the present study, we successfully prepared and characterized PanP-DM1 as a model of PDCs, which is an anti-EGFR pro-antibody following conjugation with DM1 at lysine residues. The drug-to-antibody ratio (DAR) of PanP-DM1 was determined by LC-MS after deglycosylation, and verified by UV/vis spectroscopy. Following reduction or IdeS digestion, the pro-antibody fragments linked with DM1 were investigated by middle-down mass spectrometry. Furthermore, more than 20 modified lysine conjugation sites were determined by peptide mapping after trypsin digestion. Additionally, more than ten glycoforms of PanP-DM1 were also identified and quantified. In summary, critical quality attributes (CQAs) of PDCs including DAR, drug load distribution, and conjugation sites were fully characterized, which would contribute to the development of other PDCs for cancer treatment.
Collapse
Affiliation(s)
- Boning Liu
- School of Bioscience and Bioengineering, South China University of Technology , 381 Wushan Road, Guangzhou 510641, China.,International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Huaizu Guo
- State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China.,Shanghai Zhangjiang Biotechnology Co. , 99 Libing Road, Shanghai 201203, China
| | - Junjie Zhang
- School of Bioscience and Bioengineering, South China University of Technology , 381 Wushan Road, Guangzhou 510641, China.,International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Jingya Xue
- State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China.,Shanghai Zhangjiang Biotechnology Co. , 99 Libing Road, Shanghai 201203, China.,School of Life Sciences, Fudan University , 220 Handan Road, Shanghai 200433, China
| | - Yun Yang
- State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China.,School of Life Basic Medical Sciences, Xin Xiang Medical University , 601 Jinsui Road, Xinxiang 453003, China
| | - Ting Qin
- School of Bioscience and Bioengineering, South China University of Technology , 381 Wushan Road, Guangzhou 510641, China.,International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Jin Xu
- State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China.,Shanghai Zhangjiang Biotechnology Co. , 99 Libing Road, Shanghai 201203, China
| | - Qingcheng Guo
- International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Dapeng Zhang
- International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Weizhu Qian
- State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China.,Shanghai Zhangjiang Biotechnology Co. , 99 Libing Road, Shanghai 201203, China
| | - Bohua Li
- International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Sheng Hou
- International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Jianxin Dai
- International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China
| | - Yajun Guo
- School of Bioscience and Bioengineering, South China University of Technology , 381 Wushan Road, Guangzhou 510641, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China.,School of Pharmacy, Liaocheng University , 1 Hunan Road, Liaocheng 252000, China
| | - Hao Wang
- International Joint Cancer Institute, Second Military Medical University , 800 Xiangyin Road, Shanghai 200433, China.,State Key Laboratory of Antibody Medicine and Targeted Therapy, Shanghai Key Laboratory of Cell Engineering , 99 Libing Road, Shanghai 201203, China.,School of Pharmacy, Liaocheng University , 1 Hunan Road, Liaocheng 252000, China
| |
Collapse
|
90
|
Cook BE, Adumeau P, Membreno R, Carnazza KE, Brand C, Reiner T, Agnew BJ, Lewis JS, Zeglis BM. Pretargeted PET Imaging Using a Site-Specifically Labeled Immunoconjugate. Bioconjug Chem 2016; 27:1789-95. [PMID: 27356886 DOI: 10.1021/acs.bioconjchem.6b00235] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In recent years, both site-specific bioconjugation techniques and bioorthogonal pretargeting strategies have emerged as exciting technologies with the potential to improve the safety and efficacy of antibody-based nuclear imaging. In the work at hand, we have combined these two approaches to create a pretargeted PET imaging strategy based on the rapid and bioorthogonal inverse electron demand Diels-Alder reaction between a (64)Cu-labeled tetrazine radioligand ((64)Cu-Tz-SarAr) and a site-specifically modified huA33-trans-cyclooctene immunoconjugate ((ss)huA33-PEG12-TCO). A bioconjugation strategy that harnesses enzymatic transformations and strain-promoted azide-alkyne click chemistry was used to site-specifically append PEGylated TCO moieties to the heavy chain glycans of the colorectal cancer-targeting huA33 antibody. Preclinical in vivo validation studies were performed in athymic nude mice bearing A33 antigen-expressing SW1222 human colorectal carcinoma xenografts. To this end, mice were administered (ss)huA33-PEG12-TCO via tail vein injection and-following accumulation intervals of 24 or 48 h-(64)Cu-Tz-SarAr. PET imaging and biodistribution studies reveal that this strategy clearly delineates tumor tissue as early as 1 h post-injection (6.7 ± 1.7%ID/g at 1 h p.i.), producing images with excellent contrast and high tumor-to-background activity concentration ratios (tumor:muscle = 21.5 ± 5.6 at 24 h p.i.). Furthermore, dosimetric calculations illustrate that this pretargeting approach produces only a fraction of the overall effective dose (0.0214 mSv/MBq; 0.079 rem/mCi) of directly labeled radioimmunoconjugates. Ultimately, this method effectively facilitates the high contrast pretargeted PET imaging of colorectal carcinoma using a site-specifically modified immunoconjugate.
Collapse
Affiliation(s)
- Brendon E Cook
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , 365 Fifth Avenue, New York, New York 10016, United States
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States
| | - Rosemery Membreno
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , 365 Fifth Avenue, New York, New York 10016, United States
| | | | | | - Thomas Reiner
- Department of Radiology, Weill Cornell Medical College , 520 East 70th Street, New York, New York 10065, United States
| | - Brian J Agnew
- Licensing and Commercial Supply, Thermo Fisher Scientific , 29851 Willow Creek Road, Eugene, Oregon 97402, United States
| | - Jason S Lewis
- Department of Radiology, Weill Cornell Medical College , 520 East 70th Street, New York, New York 10065, United States
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York , 413 East 69th Street, New York, New York 10028, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York , 365 Fifth Avenue, New York, New York 10016, United States.,Department of Radiology, Weill Cornell Medical College , 520 East 70th Street, New York, New York 10065, United States
| |
Collapse
|
91
|
Mechanistic and quantitative insight into cell surface targeted molecular imaging agent design. Sci Rep 2016; 6:25424. [PMID: 27147293 PMCID: PMC4857130 DOI: 10.1038/srep25424] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/15/2016] [Indexed: 12/21/2022] Open
Abstract
Molecular imaging agent design involves simultaneously optimizing multiple probe properties. While several desired characteristics are straightforward, including high affinity and low non-specific background signal, in practice there are quantitative trade-offs between these properties. These include plasma clearance, where fast clearance lowers background signal but can reduce target uptake, and binding, where high affinity compounds sometimes suffer from lower stability or increased non-specific interactions. Further complicating probe development, many of the optimal parameters vary depending on both target tissue and imaging agent properties, making empirical approaches or previous experience difficult to translate. Here, we focus on low molecular weight compounds targeting extracellular receptors, which have some of the highest contrast values for imaging agents. We use a mechanistic approach to provide a quantitative framework for weighing trade-offs between molecules. Our results show that specific target uptake is well-described by quantitative simulations for a variety of targeting agents, whereas non-specific background signal is more difficult to predict. Two in vitro experimental methods for estimating background signal in vivo are compared – non-specific cellular uptake and plasma protein binding. Together, these data provide a quantitative method to guide probe design and focus animal work for more cost-effective and time-efficient development of molecular imaging agents.
Collapse
|
92
|
Affiliation(s)
- David Y. Jackson
- Igenica Biotherapeutics, 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| |
Collapse
|
93
|
Williams SP, Ogasawara A, Tinianow JN, Flores JE, Kan D, Lau J, Go M, Vanderbilt AN, Gill HS, Miao L, Goldsmith J, Rubinfeld B, Mao W, Firestein R, Yu SF, Marik J, Terwisscha van Scheltinga AG. ImmunoPET helps predicting the efficacy of antibody-drug conjugates targeting TENB2 and STEAP1. Oncotarget 2016; 7:25103-12. [PMID: 27029064 PMCID: PMC5041891 DOI: 10.18632/oncotarget.8390] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/04/2016] [Indexed: 11/25/2022] Open
Abstract
The efficacy of antibody-drug conjugates (ADCs) targeted to solid tumors depends on biological processes that are hard to monitor in vivo. 89Zr-immunoPET of the ADC antibodies could help understand the performance of ADCs in the clinic by confirming the necessary penetration, binding, and internalization. This work studied monomethyl auristatin E (MMAE) ADCs against two targets in metastatic castration-resistant prostate cancer, TENB2 and STEAP1, in four patient-derived tumor models (LuCaP35V, LuCaP70, LuCaP77, LuCaP96.1). Three aspects of ADC biology were measured and compared: efficacy was measured in tumor growth inhibition studies; target expression was measured by immunohistochemistry and flow cytometry; and tumor antibody uptake was measured with 111In-mAbs and gamma counting or with 89Zr-immunoPET. Within each model, the mAb with the highest tumor uptake showed the greatest potency as an ADC. Sensitivity between models varied, with the LuCaP77 model showing weak efficacy despite high target expression and high antibody uptake. Ex vivo analysis confirmed the in vivo results, showing a correlation between expression, uptake and ADC efficacy. We conclude that 89Zr-immunoPET data can demonstrate which ADC candidates achieve the penetration, binding, and internalization necessary for efficacy in tumors sensitive to the toxic payload.
Collapse
Affiliation(s)
- Simon-Peter Williams
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Annie Ogasawara
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Jeff N. Tinianow
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Judith E. Flores
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - David Kan
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Jeffrey Lau
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - MaryAnn Go
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Alexander N. Vanderbilt
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Herman S. Gill
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Li Miao
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Joshua Goldsmith
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Bonnee Rubinfeld
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Weiguang Mao
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Ron Firestein
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Shang-Fan Yu
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Jan Marik
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Anton G.T. Terwisscha van Scheltinga
- Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 94080, USA
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, 9700RB, The Netherlands
| |
Collapse
|
94
|
Kraynov E, Kamath AV, Walles M, Tarcsa E, Deslandes A, Iyer RA, Datta-Mannan A, Sriraman P, Bairlein M, Yang JJ, Barfield M, Xiao G, Escandon E, Wang W, Rock DA, Chemuturi NV, Moore DJ. Current Approaches for Absorption, Distribution, Metabolism, and Excretion Characterization of Antibody-Drug Conjugates: An Industry White Paper. Drug Metab Dispos 2016; 44:617-23. [PMID: 26669328 DOI: 10.1124/dmd.115.068049] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/14/2015] [Indexed: 11/22/2022] Open
Abstract
An antibody-drug conjugate (ADC) is a unique therapeutic modality composed of a highly potent drug molecule conjugated to a monoclonal antibody. As the number of ADCs in various stages of nonclinical and clinical development has been increasing, pharmaceutical companies have been exploring diverse approaches to understanding the disposition of ADCs. To identify the key absorption, distribution, metabolism, and excretion (ADME) issues worth examining when developing an ADC and to find optimal scientifically based approaches to evaluate ADC ADME, the International Consortium for Innovation and Quality in Pharmaceutical Development launched an ADC ADME working group in early 2014. This white paper contains observations from the working group and provides an initial framework on issues and approaches to consider when evaluating the ADME of ADCs.
Collapse
Affiliation(s)
- Eugenia Kraynov
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Amrita V Kamath
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Markus Walles
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Edit Tarcsa
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Antoine Deslandes
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Ramaswamy A Iyer
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Amita Datta-Mannan
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Priya Sriraman
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Michaela Bairlein
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Johnny J Yang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Matthew Barfield
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Guangqing Xiao
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Enrique Escandon
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Weirong Wang
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Dan A Rock
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - Nagendra V Chemuturi
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| | - David J Moore
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer Inc., La Jolla, California (E.K.); Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, South San Francisco, California (A.V.K.); Drug Metabolism and Pharmacokinetics, Novartis Institutes for BioMedical Research, Novartis Pharma, Basel, Switzerland (M.W.); Drug Metabolism, Pharmacokinetics, and Bioanalysis Department, AbbVie, Worcester, Massachusetts (E.T.); Disposition, Safety and Animal Research, Sanofi, Vitry sur Seine, France (A.D.); Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (R.A.I.); Departments of Drug Disposition, Development, and Commercialization, Eli Lilly and Co., Indianapolis, Indiana (A.D.-M.); Drug Metabolism and Pharmacokinetics, Celgene Corp., Summit, New Jersey (P.S.); Drug Metabolism and Pharmacokinetics, Bayer Pharma AG, Wuppertal, Germany (Mi.B.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Boston, Massachusetts (J.J.Y.); Bioanalytical Science and Toxicokinetics, Drug Metabolism and Pharmacokinetics, GlaxoSmithKline R&D, Ware, United Kingdom (Ma.B.); Preclinical Pharmacokinetics and In Vitro ADME, Biogen, Cambridge, Massachusetts (G.X.); Biologics Discovery Drug Metabolism and Pharmacokinetics and Bioanalytics Department, Merck Research Laboratories, Palo Alto, California (E.E.); Biologics Clinical Pharmacology, Janssen R&D, Spring House, Pennsylvania, (W.W.); Amgen Pharmacokinetics and Drug Metabolism, Thousand Oaks, California (D.A.R.); Seattle Genetics Inc., Seattle, Washington (N.V.C); and Department of Pharmaceutical Sciences, Roche Innovation Center, New York City, New York (D.J.M.)
| |
Collapse
|
95
|
Tibbitts J, Canter D, Graff R, Smith A, Khawli LA. Key factors influencing ADME properties of therapeutic proteins: A need for ADME characterization in drug discovery and development. MAbs 2015; 8:229-45. [PMID: 26636901 PMCID: PMC4966629 DOI: 10.1080/19420862.2015.1115937] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein therapeutics represent a diverse array of biologics including antibodies, fusion proteins, and therapeutic replacement enzymes. Since their inception, they have revolutionized the treatment of a wide range of diseases including respiratory, vascular, autoimmune, inflammatory, infectious, and neurodegenerative diseases, as well as cancer. While in vivo pharmacokinetic, pharmacodynamic, and efficacy studies are routinely carried out for protein therapeutics, studies that identify key factors governing their absorption, distribution, metabolism, and excretion (ADME) properties have not been fully investigated. Thorough characterization and in-depth study of their ADME properties are critical in order to support drug discovery and development processes for the production of safer and more effective biotherapeutics. In this review, we discuss the main factors affecting the ADME characteristics of these large macromolecular therapies. We also give an overview of the current tools, technologies, and approaches available to investigate key factors that influence the ADME of recombinant biotherapeutic drugs, and demonstrate how ADME studies will facilitate their future development.
Collapse
|
96
|
Gordon MR, Canakci M, Li L, Zhuang J, Osborne B, Thayumanavan S. Field Guide to Challenges and Opportunities in Antibody-Drug Conjugates for Chemists. Bioconjug Chem 2015; 26:2198-215. [PMID: 26308881 PMCID: PMC4933296 DOI: 10.1021/acs.bioconjchem.5b00399] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Antibody-drug conjugates have attracted a great amount of attention as a therapeutic strategy for diseases where targeting specific tissues and cells are critical components, such as in cancer therapy. Although promising, the number of approved ADC drugs is relatively limited. This emanates from the challenges associated with generating the conjugates and the complexities associated with the stability requirements for these conjugates during circulation and after reaching the target. Here, we provide a comprehensive overview of the design challenges facing the ADC field. These challenges also provide several unique research and development opportunities, which are also highlighted throughout the review.
Collapse
Affiliation(s)
- Mallory R. Gordon
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
| | - Mine Canakci
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003 (USA)
| | - Longyu Li
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
| | - Jiaming Zhuang
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
| | - Barbara Osborne
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003 (USA)
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, MA 01003 (USA)
| | - S. Thayumanavan
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003 (USA)
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003 (USA)
| |
Collapse
|
97
|
Siegmund V, Schmelz S, Dickgiesser S, Beck J, Ebenig A, Fittler H, Frauendorf H, Piater B, Betz UAK, Avrutina O, Scrima A, Fuchsbauer H, Kolmar H. Locked by Design: A Conformationally Constrained Transglutaminase Tag Enables Efficient Site‐Specific Conjugation. Angew Chem Int Ed Engl 2015; 54:13420-4. [DOI: 10.1002/anie.201504851] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Vanessa Siegmund
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Stefan Schmelz
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Inhoffenstr. 7, 38124 Braunschweig (Germany)
| | - Stephan Dickgiesser
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Jan Beck
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Aileen Ebenig
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Heiko Fittler
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Holm Frauendorf
- Institut für Organische und Biomolekulare Chemie, Zentrale Analytik/Massenspektrometrie, Georg‐August‐Universität Göttingen, Tammannstr. 2, 37077 Göttingen (Germany)
| | - Birgit Piater
- Merck KGaA, Frankfurterstr. 250, 64293 Darmstadt (Germany)
| | | | - Olga Avrutina
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Andrea Scrima
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Inhoffenstr. 7, 38124 Braunschweig (Germany)
| | - Hans‐Lothar Fuchsbauer
- Fachbereich Chemie‐ und Biotechnologie, Hochschule Darmstadt, Schnittspahnstraße 12, 64287 Darmstadt (Germany)
| | - Harald Kolmar
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| |
Collapse
|
98
|
A Humanized Anti-CD22-Onconase Antibody-Drug Conjugate Mediates Highly Potent Destruction of Targeted Tumor Cells. J Immunol Res 2015; 2015:561814. [PMID: 26605343 PMCID: PMC4641194 DOI: 10.1155/2015/561814] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 10/01/2015] [Indexed: 11/25/2022] Open
Abstract
Antibody-drug conjugates (ADCs) have evolved as a new class of potent cancer therapeutics. We here report on the development of ADCs with specificity for the B-cell lineage specific (surface) antigen CD22 being expressed in the majority of hematological malignancies. As targeting moiety a previously generated humanized anti-CD22 single-chain variable fragment (scFv) derivative from the monoclonal antibody RFB4 was reengineered into a humanized IgG1 antibody format (huRFB4). Onconase (ranpirnase), a clinically active pancreatic-type ribonuclease, was employed as cytotoxic payload moiety. Chemical conjugation via thiol-cleavable disulfide linkage retained full enzymatic activity and full binding affinity of the ADC. Development of sophisticated purification procedures using size exclusion and ion exchange chromatography allowed the separation of immunoconjugate species with stoichiometrically defined number of Onconase cargos. A minimum of two Onconase molecules per IgG was required for achieving significant in vitro cytotoxicity towards lymphoma and leukemia cell lines. Antibody-drug conjugates with an Onconase to antibody ratio of 3 : 1 exhibited an IC50 of 0.08 nM, corresponding to more than 18,400-fold increased cytotoxicity of the ADC when compared with unconjugated Onconase. These results justify further development of this ADC as a promising first-in-class compound for the treatment of CD22-positive malignancies.
Collapse
|
99
|
Behrens CR, Ha EH, Chinn LL, Bowers S, Probst G, Fitch-Bruhns M, Monteon J, Valdiosera A, Bermudez A, Liao-Chan S, Wong T, Melnick J, Theunissen JW, Flory MR, Houser D, Venstrom K, Levashova Z, Sauer P, Migone TS, van der Horst EH, Halcomb RL, Jackson DY. Antibody-Drug Conjugates (ADCs) Derived from Interchain Cysteine Cross-Linking Demonstrate Improved Homogeneity and Other Pharmacological Properties over Conventional Heterogeneous ADCs. Mol Pharm 2015; 12:3986-98. [PMID: 26393951 DOI: 10.1021/acs.molpharmaceut.5b00432] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Conventional antibody-drug conjugates (ADCs) are heterogeneous mixtures of chemically distinct molecules that vary in both drugs/antibody (DAR) and conjugation sites. Suboptimal properties of heterogeneous ADCs have led to new site-specific conjugation methods for improving ADC homogeneity. Most site-specific methods require extensive antibody engineering to identify optimal conjugation sites and introduce unique functional groups for conjugation with appropriately modified linkers. Alternative nonrecombinant methods have emerged in which bifunctional linkers are utilized to cross-link antibody interchain cysteines and afford ADCs containing four drugs/antibody. Although these methods have been shown to improve ADC homogeneity and stability in vitro, their effect on the pharmacological properties of ADCs in vivo is unknown. In order to determine the relative impact of interchain cysteine cross-linking on the therapeutic window and other properties of ADCs in vivo, we synthesized a derivative of the known ADC payload, MC-MMAF, that contains a bifunctional dibromomaleimide (DBM) linker instead of a conventional maleimide (MC) linker. The DBM-MMAF derivative was conjugated to trastuzumab and a novel anti-CD98 antibody to afford ADCs containing predominantly four drugs/antibody. The pharmacological properties of the resulting cross-linked ADCs were compared with analogous heterogeneous ADCs derived from conventional linkers. The results demonstrate that DBM linkers can be applied directly to native antibodies, without antibody engineering, to yield highly homogeneous ADCs via cysteine cross-linking. The resulting ADCs demonstrate improved pharmacokinetics, superior efficacy, and reduced toxicity in vivo compared to analogous conventional heterogeneous ADCs.
Collapse
Affiliation(s)
- Christopher R Behrens
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Edward H Ha
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Lawrence L Chinn
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Simeon Bowers
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Gary Probst
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Maureen Fitch-Bruhns
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Jorge Monteon
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Amanda Valdiosera
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Abel Bermudez
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Sindy Liao-Chan
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Tiffany Wong
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Jonathan Melnick
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Jan-Willem Theunissen
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Mark R Flory
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Derrick Houser
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Kristy Venstrom
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Zoia Levashova
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Paul Sauer
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Thi-Sau Migone
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Edward H van der Horst
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - Randall L Halcomb
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| | - David Y Jackson
- Igenica Biotherapeutics , 863A Mitten Road, Suite 100B, Burlingame, California 94010, United States
| |
Collapse
|
100
|
Siegmund V, Schmelz S, Dickgiesser S, Beck J, Ebenig A, Fittler H, Frauendorf H, Piater B, Betz UAK, Avrutina O, Scrima A, Fuchsbauer H, Kolmar H. Durch Design verbrückt: ein konformativ eingeschränkter Transglutaminase‐Marker ermöglicht effiziente ortsspezifische Konjugation. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201504851] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Vanessa Siegmund
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Stefan Schmelz
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Braunschweig (Deutschland)
| | - Stephan Dickgiesser
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Jan Beck
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Aileen Ebenig
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Heiko Fittler
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Holm Frauendorf
- Institut für Organische und Biomolekulare Chemie, Zentrale Analytik/Massenspektrometrie, Universität Göttingen (Deutschland)
| | | | | | - Olga Avrutina
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Andrea Scrima
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Braunschweig (Deutschland)
| | | | - Harald Kolmar
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| |
Collapse
|