1
|
Khera E, Dharmarajan L, Hainzl D, Engelhardt V, Vostiarova H, Davis J, Ebel N, Wuersch K, Romanet V, Sharaby S, Kearns JD. QSP modeling of a transiently inactivating antibody-drug conjugate highlights benefit of short antibody half life. J Pharmacokinet Pharmacodyn 2024; 52:7. [PMID: 39690276 DOI: 10.1007/s10928-024-09956-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/19/2024]
Abstract
Antibody drug conjugates (ADC) are a promising class of oncology therapeutics consisting of an antibody conjugated to a payload via a linker. DYP688 is a novel ADC comprising of a signaling protein inhibitor payload (FR900359) that undergoes unique on-antibody inactivation in plasma, resulting in complex pharmacology. To assess the impact of FR inactivation on DYP688 pharmacology and clinical developability, we performed translational modeling of preclinical PK and tumor growth inhibition (TGI) data, accompanied by mechanistic Krogh cylinder tumor modeling. Using a PK-TGI model, we identified a composite exposure-above-tumorostatic concentration (AUCTSC) metric as the PK-driver of efficacy. To underpin the mechanisms behind AUCTSC as the driver of efficacy, we performed quantitative systems pharmacology (QSP) modeling of DYP688 intratumoral pharmacokinetics and pharmacodynamics. Through exploratory simulations, we show that by deviating from canonical ADC design dogma, DYP688 has optimal FR900359 activity despite its transient inactivation. Finally, we performed the successful preclinical to clinical translation of DYP688 PK, including the payload inactivation kinetics, evidenced by good agreement of the predicted PK to the observed interim clinical PK. Overall, this work highlights early quantitative pharmacokinetics as a missing link in the ADC design-developability chasm.
Collapse
Affiliation(s)
- Eshita Khera
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Lekshmi Dharmarajan
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - Dominik Hainzl
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Volker Engelhardt
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - Helena Vostiarova
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Basel, Switzerland
| | - John Davis
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA
| | - Nicolas Ebel
- Oncology, Novartis Biomedical Research, Basel, Switzerland
| | - Kuno Wuersch
- Preclinical Safety, Novartis Biomedical Research, Basel, Switzerland
| | | | - Sherif Sharaby
- PK Sciences, Translational Medicine, Novartis Biomedical Research, East Hanover, NJ, USA
| | - Jeffrey D Kearns
- PK Sciences, Translational Medicine, Novartis Biomedical Research, Cambridge, MA, USA.
| |
Collapse
|
2
|
Henise J, Hangasky JA, Charych D, Carreras CW, Ashley GW, Santi DV. A platform technology for ultra-long acting intratumoral therapy. Sci Rep 2024; 14:14000. [PMID: 38890412 PMCID: PMC11189489 DOI: 10.1038/s41598-024-64261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Intratumoral (IT) therapy is a powerful method of controlling tumor growth, but a major unsolved problem is the rapidity that injected drugs exit tumors, limiting on-target exposure and efficacy. We have developed a generic long acting IT delivery system in which a drug is covalently tethered to hydrogel microspheres (MS) by a cleavable linker; upon injection the conjugate forms a depot that slowly releases the drug and "bathes" the tumor for long periods. We established technology to measure tissue pharmacokinetics and studied MSs attached to SN-38, a topoisomerase 1 inhibitor. When MS ~ SN-38 was injected locally, tissues showed high levels of SN-38 with a long half-life of ~ 1 week. IT MS ~ SN-38 was ~ tenfold more efficacious as an anti-tumor agent than systemic SN-38. We also propose and provide an example that long-acting IT therapy might enable safe use of two drugs with overlapping toxicities. Here, long-acting IT MS ~ SN-38 is delivered with concurrent systemic PARP inhibitor. The tumor is exposed to both drugs whereas other tissues are exposed only to the systemic drug; synergistic anti-tumor activity supported the validity of this approach. We propose use of this approach to increase efficacy and reduce toxicities of combinations of immune checkpoint inhibitors such as αCTLA-4 and αPD-1.
Collapse
Affiliation(s)
- Jeff Henise
- ProLynx, 135 Mississippi Street, San Francisco, CA, 94107, USA
| | - John A Hangasky
- ProLynx, 135 Mississippi Street, San Francisco, CA, 94107, USA
| | - Deborah Charych
- Nektar, 455 Mission Bay Blvd. South, San Francisco, CA, USA
- ShynianBio Inc., 1001 17th St., San Francisco, CA, 94107, USA
| | | | - Gary W Ashley
- ProLynx, 135 Mississippi Street, San Francisco, CA, 94107, USA
| | - Daniel V Santi
- ProLynx, 135 Mississippi Street, San Francisco, CA, 94107, USA.
| |
Collapse
|
3
|
Iyer K, Ivanov J, Tenchov R, Ralhan K, Rodriguez Y, Sasso JM, Scott S, Zhou QA. Emerging Targets and Therapeutics in Immuno-Oncology: Insights from Landscape Analysis. J Med Chem 2024; 67:8519-8544. [PMID: 38787632 PMCID: PMC11181335 DOI: 10.1021/acs.jmedchem.4c00568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
In the ever-evolving landscape of cancer research, immuno-oncology stands as a beacon of hope, offering novel avenues for treatment. This study capitalizes on the vast repository of immuno-oncology-related scientific documents within the CAS Content Collection, totaling over 350,000, encompassing journals and patents. Through a pioneering approach melding natural language processing with the CAS indexing system, we unveil over 300 emerging concepts, depicted in a comprehensive "Trend Landscape Map". These concepts, spanning therapeutic targets, biomarkers, and types of cancers among others, are hierarchically organized into eight major categories. Delving deeper, our analysis furnishes detailed quantitative metrics showcasing growth trends over the past three years. Our findings not only provide valuable insights for guiding future research endeavors but also underscore the merit of tapping the vast and unparalleled breadth of existing scientific information to derive profound insights.
Collapse
Affiliation(s)
| | - Julian Ivanov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Sabina Scott
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
4
|
Vasalou C, Proia TA, Kazlauskas L, Przybyla A, Sung M, Mamidi S, Maratea K, Griffin M, Sargeant R, Urosevic J, Rosenbaum AI, Yuan J, Aluri KC, Ramsden D, Hariparsad N, Jones RD, Mettetal JT. Quantitative evaluation of trastuzumab deruxtecan pharmacokinetics and pharmacodynamics in mouse models of varying degrees of HER2 expression. CPT Pharmacometrics Syst Pharmacol 2024; 13:994-1005. [PMID: 38532525 PMCID: PMC11179703 DOI: 10.1002/psp4.13133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Trastuzumab deruxtecan (T-DXd; DS-8201; ENHERTU®) is a human epithelial growth factor receptor 2 (HER2)-directed antibody drug conjugate (ADC) with demonstrated antitumor activity against a range of tumor types. Aiming to understand the relationship between antigen expression and downstream efficacy outcomes, T-DXd was administered in tumor-bearing mice carrying NCI-N87, Capan-1, JIMT-1, and MDA-MB-468 xenografts, characterized by varying HER2 levels. Plasma pharmacokinetics (PK) of total antibody, T-DXd, and released DXd and tumor concentrations of released DXd were evaluated, in addition to monitoring γΗ2AX and pRAD50 pharmacodynamic (PD) response. A positive relationship was observed between released DXd concentrations in tumor and HER2 expression, with NCI-N87 xenografts characterized by the highest exposures compared to the remaining cell lines. γΗ2AX and pRAD50 demonstrated a sustained increase over several days occurring with a time delay relative to tumoral-released DXd concentrations. In vitro investigations of cell-based DXd disposition facilitated the characterization of DXd kinetics across tumor cells. These outputs were incorporated into a mechanistic mathematical model, utilized to describe PK/PD trends. The model captured plasma PK across dosing arms as well as tumor PK in NCI-N87, Capan-1, and MDA-MB-468 models; tumor concentrations in JIMT-1 xenografts required additional parameter adjustments reflective of complex receptor dynamics. γΗ2AX longitudinal trends were well characterized via a unified PD model implemented across xenografts demonstrating the robustness of measured PD trends. This work supports the application of a mechanistic model as a quantitative tool, reliably projecting tumor payload concentrations upon T-DXd administration, as the first step towards preclinical-to-clinical translation.
Collapse
Affiliation(s)
| | | | | | - Anna Przybyla
- AstraZeneca Research & DevelopmentWalthamMassachusettsUSA
| | - Matthew Sung
- AstraZeneca Research & DevelopmentWalthamMassachusettsUSA
| | | | - Kim Maratea
- Clinical Pharmacology & Safety SciencesWalthamMassachusettsUSA
| | - Matthew Griffin
- Clinical Pharmacology & Safety SciencesWalthamMassachusettsUSA
| | | | | | - Anton I. Rosenbaum
- Integrated Bioanalysis, Clinical Pharmacology & Safety SciencesSouth San FranciscoCaliforniaUSA
| | - Jiaqi Yuan
- Integrated Bioanalysis, Clinical Pharmacology & Safety SciencesSouth San FranciscoCaliforniaUSA
| | | | - Diane Ramsden
- AstraZeneca Research & DevelopmentWalthamMassachusettsUSA
| | | | | | | |
Collapse
|
5
|
Lami I, Wiemer AJ. Antibody-Drug Conjugates in the Pipeline for Treatment of Melanoma: Target and Pharmacokinetic Considerations. Drugs R D 2024; 24:129-144. [PMID: 38951479 PMCID: PMC11315830 DOI: 10.1007/s40268-024-00473-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/03/2024] Open
Abstract
Melanoma is an aggressive, rapidly developing form of skin cancer that affects about 22 per 100,000 individuals. Treatment options for melanoma patients are limited and typically involve surgical excision of moles and chemotherapy. Survival has been improved in recent years through targeted small molecule inhibitors and antibody-based immunotherapies. However, the long-term side effects that arise from taking chemotherapies can negatively impact the lives of patients because they lack specificity and impact healthy cells along with the cancer cells. Antibody-drug conjugates are a promising new class of drugs for the treatment of melanoma. This review focuses on the development of antibody-drug conjugates for melanoma and discusses the existing clinical trials of antibody-drug conjugates and their use as a melanoma treatment. So far, the antibody-drug conjugates have struggled from efficacy problems, with modest effects at best, leading many to be discontinued for melanoma. At the same time, conjugates such as AMT-253, targeting melanoma cell adhesion molecule, and mecbotamab vedotin targeting AXL receptor tyrosine kinase, are among the most exciting for melanoma treatment in the future.
Collapse
Affiliation(s)
- Ina Lami
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT, 06269, USA
| | - Andrew J Wiemer
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, 69 N Eagleville Road, Storrs, CT, 06269, USA.
| |
Collapse
|
6
|
Rubahamya B, Dong S, Thurber GM. Clinical translation of antibody drug conjugate dosing in solid tumors from preclinical mouse data. SCIENCE ADVANCES 2024; 10:eadk1894. [PMID: 38820153 PMCID: PMC11141632 DOI: 10.1126/sciadv.adk1894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Antibody drug conjugates (ADCs) have made impressive strides in the clinic in recent years with 11 Food and Drug Administration approvals, including 6 for the treatment of patients with solid tumors. Despite this success, the development of new agents remains challenging with a high failure rate in the clinic. Here, we show that current approved ADCs for the treatment of patients with solid tumors can all show substantial efficacy in some mouse models when administered at a similar weight-based [milligrams per kilogram (mg/kg)] dosing in mice that is tolerated in the clinic. Mechanistically, equivalent mg/kg dosing results in a similar drug concentration in the tumor and a similar tissue penetration into the tumor due to the unique delivery features of ADCs. Combined with computational approaches, which can account for the complex distribution within the tumor microenvironment, these scaling concepts may aid in the evaluation of new agents and help design therapeutics with maximum clinical efficacy.
Collapse
Affiliation(s)
- Baron Rubahamya
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shujun Dong
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M. Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
8
|
Yin L, Xu A, Zhao Y, Gu J. Bioanalytical Assays for Pharmacokinetic and Biodistribution Study of Antibody-Drug Conjugates. Drug Metab Dispos 2023; 51:1324-1331. [PMID: 37290939 DOI: 10.1124/dmd.123.001313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are produced by the chemical linkage of cytotoxic agents and monoclonal antibodies. The complexity and heterogeneity of ADCs and the low concentration of cytotoxic agent released in vivo poses big challenges to their bioanalysis. Understanding the pharmacokinetic behavior, exposure-safety, and exposure-efficacy relationships of ADCs is needed for their successful development. Accurate analytical methods are required to evaluate intact ADCs, total antibody, released small molecule cytotoxins, and related metabolites. The selection of appropriate bioanalysis methods for comprehensive analysis of ADCs is mainly dependent on the properties of cytotoxic agents, the chemical linker, and the attachment sites. The quality of the information about the whole pharmacokinetic profile of ADCs has been improved due to the development and improvement of analytical strategies for detection of ADCs, such as ligand-binding assays and mass spectrometry-related techniques. In this article, we will focus on the bioanalytical assays that have been used in the pharmacokinetic study of ADCs and discuss their advantages, current limitations, and potential challenges. SIGNIFICANCE STATEMENT: This article describes bioanalysis methods which have been used in pharmacokinetic study of ADCs and discusses the advantages, disadvantages and potential challenges of these assays. This review is useful and helpful and will provide insights and reference for bioanalysis and development of ADCs.
Collapse
Affiliation(s)
- Lei Yin
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun, 130012, PR China (L.Y., A.X., Y.Z., J.G.) and School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, PR China (L.Y.)
| | - Aiyun Xu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun, 130012, PR China (L.Y., A.X., Y.Z., J.G.) and School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, PR China (L.Y.)
| | - Yumeng Zhao
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun, 130012, PR China (L.Y., A.X., Y.Z., J.G.) and School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, PR China (L.Y.)
| | - Jingkai Gu
- Research Center for Drug Metabolism, School of Life Sciences, Jilin University, Changchun, 130012, PR China (L.Y., A.X., Y.Z., J.G.) and School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, PR China (L.Y.)
| |
Collapse
|
9
|
Zafar S, Armaghan M, Khan K, Hassan N, Sharifi-Rad J, Habtemariam S, Kieliszek M, Butnariu M, Bagiu IC, Bagiu RV, Cho WC. New insights into the anticancer therapeutic potential of maytansine and its derivatives. Biomed Pharmacother 2023; 165:115039. [PMID: 37364476 DOI: 10.1016/j.biopha.2023.115039] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Maytansine is a pharmacologically active 19-membered ansamacrolide derived from various medicinal plants and microorganisms. Among the most studied pharmacological activities of maytansine over the past few decades are anticancer and anti-bacterial effects. The anticancer mechanism of action is primarily mediated through interaction with the tubulin thereby inhibiting the assembly of microtubules. This ultimately leads to decreased stability of microtubule dynamics and cause cell cycle arrest, resulting in apoptosis. Despite its potent pharmacological effects, the therapeutic applications of maytansine in clinical medicine are quite limited due to its non-selective cytotoxicity. To overcome these limitations, several derivatives have been designed and developed mostly by modifying the parent structural skeleton of maytansine. These structural derivatives exhibit improved pharmacological activities as compared to maytansine. The present review provides a valuable insight into maytansine and its synthetic derivatives as anticancer agents.
Collapse
Affiliation(s)
- Sameen Zafar
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Khushbukhat Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan.
| | - Nazia Hassan
- Department of Biochemistry, University of Agriculture Faisalabad, Pakistan
| | | | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK.
| | - Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159 C, 02-776 Warsaw, Poland.
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from Timisoara, 300645, Calea Aradului 119, Timis, Romania.
| | - Iulia-Cristina Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Multidisciplinary Research Center on Antimicrobial Resistance, Timisoara, Romania
| | - Radu Vasile Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Preventive Medicine Study Center, Timisoara, Romania
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
10
|
Fontaine SD, Carreras CW, Reid RR, Ashley GW, Santi DV. A Very Long-acting Exatecan and Its Synergism with DNA Damage Response Inhibitors. CANCER RESEARCH COMMUNICATIONS 2023; 3:908-916. [PMID: 37377899 PMCID: PMC10208276 DOI: 10.1158/2767-9764.crc-22-0517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/03/2023] [Accepted: 04/18/2023] [Indexed: 06/29/2023]
Abstract
Exatecan (Exa) is a very potent inhibitor of topoisomerase I and anticancer agent. It has been intensively studied as a single agent, a large macromolecular conjugate and as the payload component of antigen-dependent antibody-drug conjugates. The current work describes an antigen-independent conjugate of Exa with polyethylene glycol (PEG) that slowly releases free Exa. Exa was conjugated to a 4-arm 40 kDa PEG through a β-eliminative cleavable linker. Pharmacokinetic studies in mice showed that the conjugate has an apparent circulating half-life of 12 hours, which reflects a composite of both the rate of renal elimination (half-life ∼18 hours) and release of Exa (half-life ∼40 hours). Remarkably, a single low dose of 10 μmol/kg PEG-Exa-only approximately 0.2 μmol/mouse-caused complete suppression of tumor growth of BRCA1-deficient MX-1 xenografts lasting over 40 days. A single low dose of 2.5 μmol/kg PEG-Exa administered with low but efficacious doses of the PARP inhibitor talazoparib showed strong synergy and caused significant tumor regression. Furthermore, the same low, single dose of PEG-Exa administered with the ATR inhibitor VX970 at doses of the DNA damage response inhibitor that do not affect tumor growth show high tumor regression, strong synergy, and synthetic lethality. Significance A circulating conjugate that slowly releases Exa is described. It is efficacious after a single dose and synergistic with ATR and PARP inhibitors.
Collapse
|
11
|
Chang HP, Le HK, Shah DK. Pharmacokinetics and Pharmacodynamics of Antibody-Drug Conjugates Administered via Subcutaneous and Intratumoral Routes. Pharmaceutics 2023; 15:pharmaceutics15041132. [PMID: 37111619 PMCID: PMC10142912 DOI: 10.3390/pharmaceutics15041132] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
We hypothesize that different routes of administration may lead to altered pharmacokinetics/pharmacodynamics (PK/PD) behavior of antibody-drug conjugates (ADCs) and may help to improve their therapeutic index. To evaluate this hypothesis, here we performed PK/PD evaluation for an ADC administered via subcutaneous (SC) and intratumoral (IT) routes. Trastuzumab-vc-MMAE was used as the model ADC, and NCI-N87 tumor-bearing xenografts were used as the animal model. The PK of multiple ADC analytes in plasma and tumors, and the in vivo efficacy of ADC, after IV, SC, and IT administration were evaluated. A semi-mechanistic PK/PD model was developed to characterize all the PK/PD data simultaneously. In addition, local toxicity of SC-administered ADC was investigated in immunocompetent and immunodeficient mice. Intratumoral administration was found to significantly increase tumor exposure and anti-tumor activity of ADC. The PK/PD model suggested that the IT route may provide the same efficacy as the IV route at an increased dosing interval and reduced dose level. SC administration of ADC led to local toxicity and reduced efficacy, suggesting difficulty in switching from IV to SC route for some ADCs. As such, this manuscript provides unprecedented insight into the PK/PD behavior of ADCs after IT and SC administration and paves the way for clinical evaluation of these routes.
Collapse
Affiliation(s)
- Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| | - Huyen Khanh Le
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY 14241, USA
| |
Collapse
|
12
|
Beaumont K, Pike A, Davies M, Savoca A, Vasalou C, Harlfinger S, Ramsden D, Ferguson D, Hariparsad N, Jones O, McGinnity D. ADME and DMPK considerations for the discovery and development of antibody drug conjugates (ADCs). Xenobiotica 2022; 52:770-785. [PMID: 36314242 DOI: 10.1080/00498254.2022.2141667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The therapeutic concept of antibody drug conjugates (ADCs) is to selectively target tumour cells with small molecule cytotoxic drugs to maximise cell kill benefit and minimise healthy tissue toxicity.An ADC generally consists of an antibody that targets a protein on the surface of tumour cells chemically linked to a warhead small molecule cytotoxic drug.To deliver the warhead to the tumour cell, the antibody must bind to the target protein and in general be internalised into the cell. Following internalisation, the cytotoxic agent can be released in the endosomal or lysosomal compartment (via different mechanisms). Diffusion or transport out of the endosome or lysosome allows the cytotoxic drug to express its cell-killing pharmacology. Alternatively, some ADCs (e.g. EDB-ADCs) rely on extracellular cleavage releasing membrane permeable warheads.One potentially important aspect of the ADC mechanism is the 'bystander effect' whereby the cytotoxic drug released in the targeted cell can diffuse out of that cell and into other (non-target expressing) tumour cells to exert its cytotoxic effect. This is important as solid tumours tend to be heterogeneous and not all cells in a tumour will express the targeted protein.The combination of large and small molecule aspects in an ADC poses significant challenges to the disposition scientist in describing the ADME properties of the entire molecule.This article will review the ADC landscape and the ADME properties of successful ADCs, with the aim of outlining best practice and providing a perspective of how the field can further facilitate the discovery and development of these important therapeutic modalities.
Collapse
Affiliation(s)
- Kevin Beaumont
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Andy Pike
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Michael Davies
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Adriana Savoca
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Christina Vasalou
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Steffi Harlfinger
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Diane Ramsden
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Douglas Ferguson
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Niresh Hariparsad
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, AstraZeneca, Boston, MA, USA
| | - Owen Jones
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| | - Dermot McGinnity
- Drug Metabolism and Pharmacokinetics, Early Oncology Research and Development, Cambridge, UK
| |
Collapse
|
13
|
Zang R, Barth A, Wong H, Marik J, Shen J, Lade J, Grove K, Durk MR, Parrott N, Rudewicz PJ, Zhao S, Wang T, Yan Z, Zhang D. Design and Measurement of Drug Tissue Concentration Asymmetry and Tissue Exposure-Effect (Tissue PK-PD) Evaluation. J Med Chem 2022; 65:8713-8734. [PMID: 35790118 DOI: 10.1021/acs.jmedchem.2c00502] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The "free drug hypothesis" assumes that, in the absence of transporters, the steady state free plasma concentrations equal to that at the site of action that elicit pharmacologic effects. While it is important to utilize the free drug hypothesis, exceptions exist that the free plasma exposures, either at Cmax, Ctrough, and Caverage, or at other time points, cannot represent the corresponding free tissue concentrations. This "drug concentration asymmetry" in both total and free form can influence drug disposition and pharmacological effects. In this review, we first discuss options to assess total and free drug concentrations in tissues. Then various drug design strategies to achieve concentration asymmetry are presented. Last, the utilities of tissue concentrations in understanding exposure-effect relationships and translational projections to humans are discussed for several therapeutic areas and modalities. A thorough understanding in plasma and tissue exposures correlation with pharmacologic effects can provide insightful guidance to aid drug discovery.
Collapse
Affiliation(s)
- Richard Zang
- IDEAYA Biosciences, South San Francisco, California 94080, United States
| | - Aline Barth
- Global Blood Therapeutics, South San Francisco, California 94080, United States
| | - Harvey Wong
- The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jan Marik
- Genentech, South San Francisco, California 98080, United States
| | - Jie Shen
- AbbVie, Irvine, California 92612, United States
| | - Julie Lade
- Amgen Inc., South San Francisco, California 94080, United States
| | - Kerri Grove
- Novartis, Emeryville, California 94608, United States
| | - Matthew R Durk
- Genentech, South San Francisco, California 98080, United States
| | - Neil Parrott
- Roche Innovation Centre, Basel CH-4070, Switzerland
| | | | | | - Tao Wang
- Coherus BioSciences, Redwood City, California 94605, United States
| | - Zhengyin Yan
- Genentech, South San Francisco, California 98080, United States
| | - Donglu Zhang
- Genentech, South San Francisco, California 98080, United States
| |
Collapse
|
14
|
In Situ Prodrug Activation by an Affibody‐Ruthenium Catalyst Hybrid for HER2‐Targeted Chemotherapy. Angew Chem Int Ed Engl 2022; 61:e202202855. [DOI: 10.1002/anie.202202855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Indexed: 11/07/2022]
|
15
|
In Situ Prodrug Activation by an Affibody–Ruthenium Catalyst Hybrid for HER2‐Targeted Chemotherapy. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202202855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
16
|
Boghaert ER, Cox MC, Vaidya KS. Pathophysiological and pharmacological considerations to improve the design and application of antibody-drug conjugates. Cancer Res 2022; 82:1858-1869. [PMID: 35298624 DOI: 10.1158/0008-5472.can-21-3236] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/01/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) have emerged as one of the pillars of clinical disease management in oncology. The biggest hurdle to widespread development and application of ADCs has been a narrow therapeutic index. Advances in antibody technologies and formats as well as novel linker and payload chemistries have begun to facilitate structural improvements to ADCs. However, the interplay of structural characteristics with physiologic and pharmacologic factors determining therapeutic success has garnered less attention. This review elaborates on the pharmacology of ADCs, the pathophysiology of cancerous tissues, and the reciprocal consequences on ADC properties and functions. While most currently approved ADCs utilize either microtubule inhibition or DNA damage as primary mechanisms of action, we present arguments to expand this repertoire and highlight the need for payload mechanisms that exploit disease-specific vulnerabilities. We promote the idea that the choice of antibody format, targeting antigen, linker properties, and payload of an ADC should be deliberately fit for purpose by taking the pathophysiology of disease and the specific pharmacology of the drug entity into account, thus allowing a higher probability of clinical success.
Collapse
Affiliation(s)
| | - Megan C Cox
- Abbvie, Inc., North Chicago, IL, United States
| | - Kedar S Vaidya
- Jazz Pharmaceuticals (United States), Palo Alto, CA, United States
| |
Collapse
|
17
|
Su Z, Xiao D, Xie F, Liu L, Wang Y, Fan S, Zhou X, Li S. Antibody-drug conjugates: Recent advances in linker chemistry. Acta Pharm Sin B 2021; 11:3889-3907. [PMID: 35024314 PMCID: PMC8727783 DOI: 10.1016/j.apsb.2021.03.042] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/17/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Antibody–drug conjugates (ADCs) are gradually revolutionizing clinical cancer therapy. The antibody–drug conjugate linker molecule determines both the efficacy and the adverse effects, and so has a major influence on the fate of ADCs. An ideal linker should be stable in the circulatory system and release the cytotoxic payload specifically in the tumor. However, existing linkers often release payloads nonspecifically and inevitably lead to off-target toxicity. This defect is becoming an increasingly important factor that restricts the development of ADCs. The pursuit of ADCs with optimal therapeutic windows has resulted in remarkable progress in the discovery and development of novel linkers. The present review summarizes the advance of the chemical trigger, linker‒antibody attachment and linker‒payload attachment over the last 5 years, and describes the ADMET properties of ADCs. This work also helps clarify future developmental directions for the linkers.
Collapse
Affiliation(s)
- Zheng Su
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Dian Xiao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fei Xie
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Lianqi Liu
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yanming Wang
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shiyong Fan
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Corresponding author. Tel: +86 10 66930603 (Shiyong Fan), +86 10 66930673 (Xinbo Zhou).
| | - Xinbo Zhou
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Corresponding author. Tel: +86 10 66930603 (Shiyong Fan), +86 10 66930673 (Xinbo Zhou).
| | - Song Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
18
|
Nessler I, Menezes B, Thurber GM. Key metrics to expanding the pipeline of successful antibody-drug conjugates. Trends Pharmacol Sci 2021; 42:803-812. [PMID: 34456094 DOI: 10.1016/j.tips.2021.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/22/2021] [Accepted: 07/29/2021] [Indexed: 01/18/2023]
Abstract
Although the recent FDA approval of six new antibody-drug conjugates (ADCs) is promising, attrition of ADCs during clinical development remains high. The inherent complexity of ADCs is a double-edged sword that provides opportunities for perfecting therapeutic action while also increasing confounding factors in therapeutic failures. ADC design drives their pharmacokinetics and pharmacodynamics, and requires deeper analysis than the commonly used Cmax and area under the curve (AUC) metrics to scale dosing to the clinic. Common features of current FDA-approved ADCs targeting solid tumors include humanized IgG1 antibody domains, highly expressed tumor receptors, and large antibody doses. The potential consequences of these shared features for clinical pharmacokinetics and mechanism of action are discussed, and key design aspects for successful solid tumor ADCs are highlighted.
Collapse
Affiliation(s)
- Ian Nessler
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bruna Menezes
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
19
|
Su D, Zhang D. Linker Design Impacts Antibody-Drug Conjugate Pharmacokinetics and Efficacy via Modulating the Stability and Payload Release Efficiency. Front Pharmacol 2021; 12:687926. [PMID: 34248637 PMCID: PMC8262647 DOI: 10.3389/fphar.2021.687926] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023] Open
Abstract
The development of antibody-drug conjugates (ADCs) has significantly been advanced in the past decade given the improvement of payloads, linkers and conjugation methods. In particular, linker design plays a critical role in modulating ADC stability in the systemic circulation and payload release efficiency in the tumors, which thus affects ADC pharmacokinetic (PK), efficacy and toxicity profiles. Previously, we have investigated key linker parameters such as conjugation chemistry (e.g., maleimide vs. disulfide), linker length and linker steric hindrance and their impacts on PK and efficacy profiles. Herein, we discuss our perspectives on development of integrated strategies for linker design to achieve a balance between ADC stability and payload release efficiency for desired efficacy in antigen-expressing xenograft models. The strategies have been successfully applied to the design of site-specific THIOMABTM antibody-drug conjugates (TDCs) with different payloads. We also propose to conduct dose fractionation studies to gain guidance for optimal dosing regimens of ADCs in pre-clinical models.
Collapse
Affiliation(s)
- Dian Su
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, United States
| | - Donglu Zhang
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
20
|
Gallo F, Korsak B, Müller C, Hechler T, Yanakieva D, Avrutina O, Kolmar H, Pahl A. Enhancing the Pharmacokinetics and Antitumor Activity of an α-Amanitin-Based Small-Molecule Drug Conjugate via Conjugation with an Fc Domain. J Med Chem 2021; 64:4117-4129. [PMID: 33755471 DOI: 10.1021/acs.jmedchem.1c00003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Herein we describe the design and biological evaluation of a novel antitumor therapeutic platform that combines the most favorable properties of small-molecule drug conjugates (SMDCs) and antibody drug conjugates (ADCs). Although the small size of SMDCs, compared to ADCs, is an appealing feature for their application in the treatment of solid tumors, SMDCs usually suffer from poor pharmacokinetics, which severely limits their therapeutic efficacy. To overcome this limitation, in this proof-of-concept study we grafted an α-amanitin-based SMDC that targets prostate cancer cells onto an immunoglobulin Fc domain via a two-step "program and arm" chemoenzymatic strategy. We demonstrated the superior pharmacokinetic properties and therapeutic efficacy of the resulting Fc-SMDC over the SMDC in a prostate cancer xenograft mouse model. This approach may provide a general strategy toward effective antitumor therapeutics combining small size with pharmacokinetic properties close to those of an ADC.
Collapse
Affiliation(s)
- Francesca Gallo
- Heidelberg Pharma Research GmbH, Heidelberg Pharma AG, Schriesheimer Str. 101, 68526 Ladenburg, Germany
| | - Barbara Korsak
- Heidelberg Pharma Research GmbH, Heidelberg Pharma AG, Schriesheimer Str. 101, 68526 Ladenburg, Germany
| | - Christoph Müller
- Heidelberg Pharma Research GmbH, Heidelberg Pharma AG, Schriesheimer Str. 101, 68526 Ladenburg, Germany
| | - Torsten Hechler
- Heidelberg Pharma Research GmbH, Heidelberg Pharma AG, Schriesheimer Str. 101, 68526 Ladenburg, Germany
| | - Desislava Yanakieva
- Department of Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Olga Avrutina
- Department of Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Harald Kolmar
- Department of Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Straße 4, 64287 Darmstadt, Germany
| | - Andreas Pahl
- Heidelberg Pharma Research GmbH, Heidelberg Pharma AG, Schriesheimer Str. 101, 68526 Ladenburg, Germany
| |
Collapse
|
21
|
Targeting Vesicular LGALS3BP by an Antibody-Drug Conjugate as Novel Therapeutic Strategy for Neuroblastoma. Cancers (Basel) 2020; 12:cancers12102989. [PMID: 33076448 PMCID: PMC7650653 DOI: 10.3390/cancers12102989] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Antibody Drug Conjugates are an emerging class of biopharmaceuticals that have seen an impressive increase of attention in the field of cancer therapy. Here, we describe the therapeutic activity of 1959-sss/DM3, a non-internalizing ADC targeting LGALS3BP, a secreted, extracellular vesicles-associated protein expressed by the majority of human cancers, including neuroblastoma. We show that 1959-sss/DM3 treatment can cure mice with established neuroblastoma tumours in pseudometastatic, orthotopic and Patient Derived Xenograft models. Abstract Neuroblastoma is the most common extra-cranial solid tumor in infants and children, which accounts for approximately 15% of all cancer-related deaths in the pediatric population. New therapeutic modalities are urgently needed. Antibody-Drug Conjugates (ADC)s-based therapy has been proposed as potential strategy to treat this pediatric malignancy. LGALS3BP is a highly glycosylated protein involved in tumor growth and progression. Studies have shown that LGALS3BP is enriched in extracellular vesicles (EV)s derived by most neuroblastoma cells, where it plays a critical role in preparing a favorable tumor microenvironment (TME) through direct cross talk between cancer and stroma cells. Here, we describe the development of a non-internalizing LGALS3BP ADC, named 1959-sss/DM3, which selectively targets LGALS3BP expressing neuroblastoma. 1959-sss/DM3 mediated potent therapeutic activity in different types of neuroblastoma models. Notably, we found that treatments were well tolerated at efficacious doses that were fully curative. These results offer preclinical proof-of-concept for an ADC targeting exosomal LGALS3BP approach for neuroblastomas.
Collapse
|
22
|
Oliveira BL, Stenton BJ, Unnikrishnan VB, de Almeida CR, Conde J, Negrão M, Schneider FSS, Cordeiro C, Ferreira MG, Caramori GF, Domingos JB, Fior R, Bernardes GJL. Platinum-Triggered Bond-Cleavage of Pentynoyl Amide and N-Propargyl Handles for Drug-Activation. J Am Chem Soc 2020; 142:10869-10880. [PMID: 32456416 PMCID: PMC7304066 DOI: 10.1021/jacs.0c01622] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
The
ability to create ways to control drug activation at specific
tissues while sparing healthy tissues remains a major challenge. The
administration of exogenous target-specific triggers offers the potential
for traceless release of active drugs on tumor sites from antibody–drug
conjugates (ADCs) and caged prodrugs. We have developed a metal-mediated
bond-cleavage reaction that uses platinum complexes [K2PtCl4 or Cisplatin (CisPt)] for drug activation. Key to
the success of the reaction is a water-promoted activation process
that triggers the reactivity of the platinum complexes. Under these
conditions, the decaging of pentynoyl tertiary amides and N-propargyls occurs rapidly in aqueous systems. In cells,
the protected analogues of cytotoxic drugs 5-fluorouracil (5-FU) and
monomethyl auristatin E (MMAE) are partially activated by nontoxic
amounts of platinum salts. Additionally, a noninternalizing ADC built
with a pentynoyl traceless linker that features a tertiary amide protected
MMAE was also decaged in the presence of platinum salts for extracellular
drug release in cancer cells. Finally, CisPt-mediated prodrug activation
of a propargyl derivative of 5-FU was shown in a colorectal zebrafish
xenograft model that led to significant reductions in tumor size.
Overall, our results reveal a new metal-based cleavable reaction that
expands the application of platinum complexes beyond those in catalysis
and cancer therapy.
Collapse
Affiliation(s)
- Bruno L Oliveira
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Benjamin J Stenton
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - V B Unnikrishnan
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Cátia Rebelo de Almeida
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal
| | - João Conde
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Magda Negrão
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal
| | - Felipe S S Schneider
- Department of Chemistry, Federal University of Santa Catarina-UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Carlos Cordeiro
- Laboratório de FT-ICR e Espectrometria de Massa Estrutural, Faculdade de Ciências da Universidade de Lisboa, Campo-Grande, 1749-016 Lisboa, Portugal
| | - Miguel Godinho Ferreira
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal.,Institute for Research on Cancer and Aging of Nice (IRCAN), Université Côte d'Azur, UMR7284 U1081 UNS, 06107 Nice, France
| | - Giovanni F Caramori
- Department of Chemistry, Federal University of Santa Catarina-UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Josiel B Domingos
- Department of Chemistry, Federal University of Santa Catarina-UFSC, Campus Trindade, Florianópolis, Santa Catarina 88040-900, Brazil
| | - Rita Fior
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Avenida Brasilia, 1400-038 Lisboa, Portugal
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
23
|
Kosaka M, Zhang D, Wong S, Yan Z. NADPH-Independent Inactivation of CYP2B6 and NADPH-Dependent Inactivation of CYP3A4/5 by PBD: Potential Implication for Assessing Covalent Modulators for Time-Dependent Inhibition. Drug Metab Dispos 2020; 48:655-661. [DOI: 10.1124/dmd.120.090878] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/13/2020] [Indexed: 02/03/2023] Open
|
24
|
Rock BM, Foti RS. Pharmacokinetic and Drug Metabolism Properties of Novel Therapeutic Modalities. Drug Metab Dispos 2019; 47:1097-1099. [PMID: 31399505 DOI: 10.1124/dmd.119.088708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022] Open
Abstract
The discovery and development of novel pharmaceutical therapies is rapidly transitioning from a small molecule-dominated focus to a more balanced portfolio consisting of small molecules, monoclonal antibodies, engineered proteins (modified endogenous proteins, bispecific antibodies, and fusion proteins), oligonucleotides, and gene-based therapies. This commentary, and the special issue as a whole, aims to highlight these emerging modalities and the efforts underway to better understand their unique pharmacokinetic and absorption, disposition, metabolism, and excretion (ADME) properties. The articles highlighted herein can be broadly grouped into those focusing on the ADME properties of novel therapeutics, those exploring targeted-delivery strategies, and finally, those discussing oligonucleotide therapies. It is also evident that whereas the field in general continues to progress toward new and more complex molecules, a significant amount of effort is still being placed on antibody-drug conjugates. As therapeutic molecules become increasingly complex, a parallel demand for advancements in experimental and analytical tools will become increasingly evident, both to increase the speed and efficiency of identifying safe and efficacious molecules and simultaneously decreasing our dependence on in vivo studies in preclinical species. The research and commentary included in this special issue will provide researchers, clinicians, and the patients we serve more options in the ongoing fight against grievous illnesses and unmet medical needs. SIGNIFICANCE STATEMENT: Recent trends in drug discovery and development suggest a shift away from a small molecule-dominated approach to a more balanced portfolio that includes small molecules, monoclonal antibodies, engineered proteins, and gene therapies. The research presented in this special issue of Drug Metabolism and Disposition will serve to highlight advancements in the understanding of the mechanisms that govern the pharmacokinetic and drug metabolism properties of the novel therapeutic modalities.
Collapse
Affiliation(s)
- Brooke M Rock
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (B.M.R.) and Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F.)
| | - Robert S Foti
- Pharmacokinetics and Drug Metabolism, Amgen Research, South San Francisco, California (B.M.R.) and Pharmacokinetics and Drug Metabolism, Amgen Research, Cambridge, Massachusetts (R.S.F.)
| |
Collapse
|