1
|
Zhao S, Yu N, Han H, Guo S, Murthy N. Advances in acid-degradable and enzyme-cleavable linkers for drug delivery. Curr Opin Chem Biol 2024; 84:102552. [PMID: 39642424 DOI: 10.1016/j.cbpa.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/23/2024] [Accepted: 11/13/2024] [Indexed: 12/08/2024]
Abstract
Drug delivery vectors have the potential to improve the efficacy of therapeutics, including small molecules and nucleic acid-based drugs. However, challenges remain in developing linkages that enable the precise and efficient release of therapeutic cargo in response to mildly acidic environments or lysosomal enzymes. This review highlights recent advances in acid-degradable acetal/ketal and enzyme-cleavable linkages for endolysosomal release. These innovations include the developments of azido-acetal linkers with improved stability and hydrolysis kinetics, organocatalytic trans-isopropenylation for synthesizing asymmetric ketals and their applications in drug delivery, and enzyme-cleavable linkers activated by cathepsin B or β-galactosidase.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Bioengineering and Innovative Genomics Institute, University of California Berkeley, 2151 Berkeley Way, Berkeley, CA 94720, USA
| | - Na Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Hesong Han
- Department of Bioengineering and Innovative Genomics Institute, University of California Berkeley, 2151 Berkeley Way, Berkeley, CA 94720, USA
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China.
| | - Niren Murthy
- Department of Bioengineering and Innovative Genomics Institute, University of California Berkeley, 2151 Berkeley Way, Berkeley, CA 94720, USA.
| |
Collapse
|
2
|
Bitsch P, Dessin C, Bitsch S, Voss J, Becker J, Sharma P, Biyani N, Kochat H, Sewald N, Kolmar H. Evaluation of Potency and Specificity of Cryptophycin-Loaded Antibody-Drug Conjugates. Chembiochem 2024:e202400738. [PMID: 39462215 DOI: 10.1002/cbic.202400738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
An enhanced variant of the antimitotic toxin cryptophycin was conjugated to the anti-Her2 monoclonal antibody (mAb) Trastuzumab upon Michael addition. Either antibodies with freed hinge-region cysteines or THIOMAB formats with engineered cysteines in the mAbs light chain were added to a maleimide derivative of cryptophycin. These Antibody-Drug Conjugates (ADCs) showed retained binding to Her2 positive tumor cells and highly efficient cell killing in double-digit pM range on high Her2-expressing SK-BR-3 cells. Two ADCs (DAR 6, DAR 3) showed superior cell killing of the cell lines JIMT-1 and RT112 with medium receptor expression level in comparison with a DAR 6 MMAE ADC serving as reference. The observed cell cytotoxicity is target-dependent since no impact on cell viability was observed for low Her2-expressing MDA-MB468 cells. Particularly the DAR 3 ADC in THIOMAB format exhibiting desirable biophysical properties and high potency emerged as a promising candidate for further in vivo investigations.
Collapse
Affiliation(s)
- Peter Bitsch
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| | - Cedric Dessin
- Organic and Bioorganic Chemistry, Faculty of Chemistry, University of Bielefeld, Universitätsstraße 25, 33615, Bielefeld, Deutschland
| | - Sebastian Bitsch
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| | - Jona Voss
- Organic and Bioorganic Chemistry, Faculty of Chemistry, University of Bielefeld, Universitätsstraße 25, 33615, Bielefeld, Deutschland
| | - Janine Becker
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| | - Panna Sharma
- Lantern Pharma Inc., 1920 McKinney Ave, 7th Floor, Dallas 75201, TX, USA
| | - Neha Biyani
- Lantern Pharma Inc., 1920 McKinney Ave, 7th Floor, Dallas 75201, TX, USA
| | - Harry Kochat
- The University of Tennessee, Health Science Center, 208 S., Dudley Street, Memphis, 38163, TN, USA
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, University of Bielefeld, Universitätsstraße 25, 33615, Bielefeld, Deutschland
| | - Harald Kolmar
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
- Centre of Synthetic Biology, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| |
Collapse
|
3
|
Dessin C, Schachtsiek T, Voss J, Abel AC, Neumann B, Stammler HG, Prota AE, Sewald N. Highly Cytotoxic Cryptophycin Derivatives with Modification in Unit D for Conjugation. Angew Chem Int Ed Engl 2024:e202416210. [PMID: 39324938 DOI: 10.1002/anie.202416210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Cytotoxic payloads for drug conjugates suitable for directed tumor therapy need to be highly potent and require a functional group for conjugation with the homing device (antibody, peptide, or small molecule). Cryptophycins are cyclodepsipeptides that stand out from the realm of natural products due to their extraordinarily high cytotoxicity. However, the installation of a suitable conjugation handle without compromising the toxicity is highly challenging. The unit D, natively 2-hydroxyisocaproic acid (leucic acid), was envisaged as a promising attachment site based on structural information from X-ray analysis. A versatile, scalable and efficient synthetic route towards conjugable cryptophycins with modification in unit D was developed and an array of new cryptophycin analogues was synthesized. Several derivatives, especially those containing lipophilic groups with low steric demand such as alkylated amino groups, exhibit low picomolar cytotoxicity often combined with efficacy against multidrug-resistant tumor cells. The newly established cryptophycin analogues comprise a broad range of relevant functional groups used as conjugation handles, among them amino, hydroxy, carboxy, as well as sulfur-containing derivatives. X-ray crystallographic analysis of a tubulin-bound cryptophycin together with quantitative structure activity relationship manifested rationales for the synthesis of most potent cryptophycin derivatives and further confirmed the suitability of modifications in unit D.
Collapse
Affiliation(s)
- Cedric Dessin
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Thomas Schachtsiek
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Jona Voss
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Anne-Catherine Abel
- PSI Center for Life Sciences, Forschungsstraße 111, 5232, Villigen PSI, Switzerland
| | - Beate Neumann
- Inorganic and Structural Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Hans-Georg Stammler
- Inorganic and Structural Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Andrea E Prota
- PSI Center for Life Sciences, Forschungsstraße 111, 5232, Villigen PSI, Switzerland
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Department of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| |
Collapse
|
4
|
Plumet C, Katsakos SD, Girard M, Jamal IA, Clarhaut J, Renoux B, Opalinski I, Papot S. An Enzyme-Responsive Self-Immolative Recognition Marker for Manipulating Cell-Cell Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402278. [PMID: 38953328 PMCID: PMC11423255 DOI: 10.1002/advs.202402278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/15/2024] [Indexed: 07/04/2024]
Abstract
The development of innovative strategies for cell membranes engineering is of prime interest to explore and manipulate cell-cell interactions. Herein, an enzyme-sensitive recognition marker that can be introduced on cell surface via bioorthogonal chemistry is designed. Once functionalized in this fashion, the cells gain the ability to assemble with cell partners coated with the complementary marker through non-covalent click chemistry. The artificial cell adhesion induces natural biological processes associated with cell proximity such as inhibiting cancer cell proliferation and migration. On the other hand, the enzymatic activation of the stimuli-responsive marker triggers the disassembly of cells, thereby restoring the tumor cell proliferation and migration rates. Thus, the study shows that the ready-to-use complementary markers are valuable tools for controlling the formation and the breaking of bonds between cells, offering an easy way to investigate biological processes associated to cell proximity.
Collapse
Affiliation(s)
- Chad Plumet
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
| | - Spyridon D. Katsakos
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
| | - Mélissa Girard
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
| | - Israa Al Jamal
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
| | - Jonathan Clarhaut
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
- University Hospital of Poitiers2 rue de la MilétriePoitiers86021France
| | - Brigitte Renoux
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
| | - Isabelle Opalinski
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
| | - Sébastien Papot
- Equipe Labellisée Ligue Contre le CancerUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)University of Poitiers4 rue Michel‐Brunet, TSA 51106, Cedex 9Poitiers86073France
| |
Collapse
|
5
|
Hohnsen J, Rryci L, Obretenova D, Friedel J, Jouchaghani S, Klein A. Functionalizing Thiosemicarbazones for Covalent Conjugation. Molecules 2024; 29:3680. [PMID: 39125087 PMCID: PMC11314635 DOI: 10.3390/molecules29153680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Thiosemicarbazones (TSCs) with their modular character (thiosemicarbazides + carbonyl compound) allow broad variation of up to four substituents on the main R1R2C=N(1)-NH-C(S)-N(4)R3R4 core and are thus interesting tools for the formation of conjugates or the functionalization of nanoparticles (NPs). In this work, di-2-pyridyl ketone was introduced for the coordination of metals and 9-anthraldehyde for luminescence as R1 and R2 to TSCs. R3 and R4 substituents were varied for the formation of conjugates. Amino acids were introduced at the N4 position to produce [R1R2TSC-spacer-amino acid] conjugates. Further, functions such as phosphonic acid (R-P(O)(OH)2), D-glucose, o-hydroquinone, OH, and thiol (SH) were introduced at the N4 position producing [R1R2TSC-spacer-anchor group] conjugates for direct NP anchoring. Phenyl, cyclohexyl, benzyl, ethyl and methyl were used as spacer units. Both phenyl phosphonic acid TSC derivatives were bound on TiO2 NPs as a first example of direct NP anchoring. [R1R2TSC-spacer-end group] conjugates including OH, S-Bn (Bn = benzyl), NH-Boc (Boc = tert-butyloxycarbonyl), COOtBu, C≡CH, or N3 end groups were synthesized for potential covalent binding to functional molecules or functionalized NPs through amide, ester, or triazole functions. The synthesis of the thiosemicarbazides H2NNH-C(S)-NR3R4 starting from amines, including amino acids, SCCl2 or CS2, and hydrazine and their condensation with dipyridyl ketone and anthraldehyde led to 34 new TSC derivatives. They were synthesized in up to six steps with overall yields ranging from 10 to 85% and were characterized by a combination of nuclear magnetic resonance spectroscopy and mass spectrometry. UV-vis absorption and photoluminescence spectroscopy allowed us to easily trace the dipyridyl imine and anthracene chromophores.
Collapse
Affiliation(s)
| | | | | | | | | | - Axel Klein
- University of Cologne, Faculty of Mathematics and Natural Sciences, Department of Chemistry and Biochemistry, Institute for Inorganic and Materials Chemistry, Greinstraße 6, 50939 Koeln, Germany; (J.H.); (L.R.); (D.O.); (J.F.); (S.J.)
| |
Collapse
|
6
|
Pezacki AT, Gonciarz RL, Okamura T, Matier CD, Torrente L, Cheng K, Miller SG, Ralle M, Ward NP, DeNicola GM, Renslo AR, Chang CJ. A tandem activity-based sensing and labeling strategy reveals antioxidant response element regulation of labile iron pools. Proc Natl Acad Sci U S A 2024; 121:e2401579121. [PMID: 38968123 PMCID: PMC11252945 DOI: 10.1073/pnas.2401579121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/29/2024] [Indexed: 07/07/2024] Open
Abstract
Iron is an essential element for life owing to its ability to participate in a diverse array of oxidation-reduction reactions. However, misregulation of iron-dependent redox cycling can also produce oxidative stress, contributing to cell growth, proliferation, and death pathways underlying aging, cancer, neurodegeneration, and metabolic diseases. Fluorescent probes that selectively monitor loosely bound Fe(II) ions, termed the labile iron pool, are potentially powerful tools for studies of this metal nutrient; however, the dynamic spatiotemporal nature and potent fluorescence quenching capacity of these bioavailable metal stores pose challenges for their detection. Here, we report a tandem activity-based sensing and labeling strategy that enables imaging of labile iron pools in live cells through enhancement in cellular retention. Iron green-1 fluoromethyl (IG1-FM) reacts selectively with Fe(II) using an endoperoxide trigger to release a quinone methide dye for subsequent attachment to proximal biological nucleophiles, providing a permanent fluorescent stain at sites of elevated labile iron. IG1-FM imaging reveals that degradation of the major iron storage protein ferritin through ferritinophagy expands the labile iron pool, while activation of nuclear factor-erythroid 2-related factor 2 (NRF2) antioxidant response elements (AREs) depletes it. We further show that lung cancer cells with heightened NRF2 activation, and thus lower basal labile iron, have reduced viability when treated with an iron chelator. By connecting labile iron pools and NRF2-ARE activity to a druggable metal-dependent vulnerability in cancer, this work provides a starting point for broader investigations into the roles of transition metal and antioxidant signaling pathways in health and disease.
Collapse
Affiliation(s)
- Aidan T. Pezacki
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Ryan L. Gonciarz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Toshitaka Okamura
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Carson D. Matier
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Laura Torrente
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Ke Cheng
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Sophia G. Miller
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Martina Ralle
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Nathan P. Ward
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA94158
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA94720
| |
Collapse
|
7
|
Jiang X, Zhu L, Wei Q, Lu W, Yu J, Zhu S. Enhancing SN38 prodrug delivery using a self-immolative linker and endogenous albumin transport. J Control Release 2024; 369:622-629. [PMID: 38604383 DOI: 10.1016/j.jconrel.2024.04.019] [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: 12/18/2023] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Enhancing the delivery and release efficiency of hydroxyl agents, constrained by high pKa values and issues of release rate or unstable linkage, is a critical challenge. To address this, a self-immolative linker, composed of a modifiable p-hydroxybenzyl ether and a fast cyclization adapter (N-(ortho-hydroxyphenyl)-N-methylcarbamate) was strategically designed, for the synthesis of prodrugs. The innovative linker not only provides a side chain modification but also facilitates the rapid release of the active payloads, thereby enabling precise drug delivery. Particularly, five prodrug model compounds (J1, J2, J3, J5 and J6) were synthesized to evaluate the release rates by using β-glucuronic acid as trigger and five hydroxyl compounds as model payloads. Significantly, all prodrug model compounds could efficiently release the hydroxyl payloads under the action of β-glucuronidase, validating the robustness of the linker. And then, to assess the drug delivery and release efficiency using endogenous albumin as a transport vehicle, J1148, a SN38 prodrug modified with maleimide side chain was synthesized. Results demonstrated that J1148 covalently bound to plasma albumin through in situ Michael addition, effectively targeting the tumor microenvironment. Activated by β-glucuronidase, J1148 underwent a classical 1, 6-elimination, followed by rapid cyclization of the adapter, thereby releasing SN38. Impressively, J1148 showed excellent therapeutic efficacy against human colonic cancer xenograft model, leading to a significant reduction or even disappearance of tumors (3/6 of mice cured). These findings underscore the potential of the designed linker in the delivery system of hydroxyl agents, positioning it at the forefront of advancements in drug delivery technology.
Collapse
Affiliation(s)
- Xing Jiang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Lingyi Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Qingyu Wei
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Jiahui Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China.
| | - Shulei Zhu
- Innovation Center for AI and Drug Discovery, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China.
| |
Collapse
|
8
|
Mason M, Bisbal Lopez L, Bashiri F, Herrero A, Baron A, Bucci R, Pignataro L, Gennari C, Dal Corso A. Amine-Carbamate Self-Immolative Spacers Counterintuitively Release 3° Alcohol at Much Faster Rates than 1° Alcohol Payloads. Chembiochem 2024; 25:e202400174. [PMID: 38415320 DOI: 10.1002/cbic.202400174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 02/29/2024]
Abstract
Self-immolative (SI) spacers are degradable chemical connectors widely used in prodrugs and drug conjugates to release pharmaceutical ingredients in response to specific stimuli. Amine-carbamate SI spacers are particularly versatile, as they have been used to release different hydroxy cargos, ranging from 2° and 3° alcohols to phenols and oximes. In this work, we describe the ability of three amine-carbamate SI spacers to release three structurally similar imidazoquinoline payloads, bearing either a 1°, a 2° or a 3° alcohol as the leaving group. While the spacers showed comparable efficacy at releasing the 2° and 3° alcohols, the liberation of the 1° alcohol was much slower, unveiling a counterintuitive trend in nucleophilic acyl substitutions. The release of the 1° alcohol payload was only possible using a SI spacer bearing a pyrrolidine ring and a tertiary amine handle, which opens the way to future applications in drug delivery systems.
Collapse
Affiliation(s)
- Mattia Mason
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| | - Lydia Bisbal Lopez
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| | - Fazel Bashiri
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| | - Aurélie Herrero
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| | - Aurélien Baron
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| | - Raffaella Bucci
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via G. Venezian 21, 20133, Milan, Italy
| | - Luca Pignataro
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| | - Cesare Gennari
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| | - Alberto Dal Corso
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi, 19, I, -20133, Milan, Italy
| |
Collapse
|
9
|
Kim H, Taslakjian B, Kim S, Tirrell MV, Guler MO. Therapeutic Peptides, Proteins and their Nanostructures for Drug Delivery and Precision Medicine. Chembiochem 2024; 25:e202300831. [PMID: 38408302 DOI: 10.1002/cbic.202300831] [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: 12/08/2023] [Revised: 02/05/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Peptide and protein nanostructures with tunable structural features, multifunctionality, biocompatibility and biomolecular recognition capacity enable development of efficient targeted drug delivery tools for precision medicine applications. In this review article, we present various techniques employed for the synthesis and self-assembly of peptides and proteins into nanostructures. We discuss design strategies utilized to enhance their stability, drug-loading capacity, and controlled release properties, in addition to the mechanisms by which peptide nanostructures interact with target cells, including receptor-mediated endocytosis and cell-penetrating capabilities. We also explore the potential of peptide and protein nanostructures for precision medicine, focusing on applications in personalized therapies and disease-specific targeting for diagnostics and therapeutics in diseases such as cancer.
Collapse
Affiliation(s)
- HaRam Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Boghos Taslakjian
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Sarah Kim
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Matthew V Tirrell
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| | - Mustafa O Guler
- The Pritzker School of Molecular Engineering, The University of Chicago, 5640 S. Ellis Ave., Chicago, 60637, IL, USA
| |
Collapse
|
10
|
Saady A, Malcolm GK, Fitzpatrick MP, Pairault N, Tizzard GJ, Mohammed S, Tavassoli A, Goldup SM. A Platform Approach to Cleavable Macrocycles for the Controlled Disassembly of Mechanically Caged Molecules. Angew Chem Int Ed Engl 2024; 63:e202400344. [PMID: 38276911 DOI: 10.1002/anie.202400344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Inspired by interlocked oligonucleotides, peptides and knotted proteins, synthetic systems where a macrocycle cages a bioactive species that is "switched on" by breaking the mechanical bond have been reported. However, to date, each example uses a bespoke chemical design. Here we present a platform approach to mechanically caged structures wherein a single macrocycle precursor is diversified at a late stage to include a range of trigger units that control ring opening in response to enzymatic, chemical, or photochemical stimuli. We also demonstrate that our approach is applicable to other classes of macrocycles suitable for rotaxane and catenane formation.
Collapse
Affiliation(s)
- Abed Saady
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Georgia K Malcolm
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Matthew P Fitzpatrick
- School of Chemistry, University of Southampton, University Road, Southampton, SO17 1BJ, UK
| | - Noel Pairault
- School of Chemistry, University of Southampton, University Road, Southampton, SO17 1BJ, UK
| | - Graham J Tizzard
- School of Chemistry, University of Southampton, University Road, Southampton, SO17 1BJ, UK
| | - Soran Mohammed
- School of Chemistry, University of Southampton, University Road, Southampton, SO17 1BJ, UK
| | - Ali Tavassoli
- School of Chemistry, University of Southampton, University Road, Southampton, SO17 1BJ, UK
| | - Stephen M Goldup
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
11
|
He X, Li J, Liang X, Mao W, Deng X, Qin M, Su H, Wu H. An all-in-one tetrazine reagent for cysteine-selective labeling and bioorthogonal activable prodrug construction. Nat Commun 2024; 15:2831. [PMID: 38565562 PMCID: PMC10987521 DOI: 10.1038/s41467-024-47188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
The prodrug design strategy offers a potent solution for improving therapeutic index and expanding drug targets. However, current prodrug activation designs are mainly responsive to endogenous stimuli, resulting in unintended drug release and systemic toxicity. In this study, we introduce 3-vinyl-6-oxymethyl-tetrazine (voTz) as an all-in-one reagent for modular preparation of tetrazine-caged prodrugs and chemoselective labeling peptides to produce bioorthogonal activable peptide-prodrug conjugates. These stable prodrugs can selectively bind to target cells, facilitating cellular uptake. Subsequent bioorthogonal cleavage reactions trigger prodrug activation, significantly boosting potency against tumor cells while maintaining exceptional off-target safety for normal cells. In vivo studies demonstrate the therapeutic efficacy and safety of this prodrug design approach. Given the broad applicability of functional groups and labeling versatility with voTz, we foresee that this strategy will offer a versatile solution to enhance the therapeutic range of cytotoxic agents and facilitate the development of bioorthogonal activatable biopharmaceuticals and biomaterials.
Collapse
Affiliation(s)
- Xinyu He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinxin Liang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wuyu Mao
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinglong Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan University, Chengdu, China
| | - Meng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Su
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Haoxing Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Ahmadi M, Ritter CA, von Woedtke T, Bekeschus S, Wende K. Package delivered: folate receptor-mediated transporters in cancer therapy and diagnosis. Chem Sci 2024; 15:1966-2006. [PMID: 38332833 PMCID: PMC10848714 DOI: 10.1039/d3sc05539f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/31/2023] [Indexed: 02/10/2024] Open
Abstract
Neoplasias pose a significant threat to aging society, underscoring the urgent need to overcome the limitations of traditional chemotherapy through pioneering strategies. Targeted drug delivery is an evolving frontier in cancer therapy, aiming to enhance treatment efficacy while mitigating undesirable side effects. One promising avenue utilizes cell membrane receptors like the folate receptor to guide drug transporters precisely to malignant cells. Based on the cellular folate receptor as a cancer cell hallmark, targeted nanocarriers and small molecule-drug conjugates have been developed that comprise different (bio) chemistries and/or mechanical properties with individual advantages and challenges. Such modern folic acid-conjugated stimuli-responsive drug transporters provide systemic drug delivery and controlled release, enabling reduced dosages, circumvention of drug resistance, and diminished adverse effects. Since the drug transporters' structure-based de novo design is increasingly relevant for precision cancer remediation and diagnosis, this review seeks to collect and debate the recent approaches to deliver therapeutics or diagnostics based on folic acid conjugated Trojan Horses and to facilitate the understanding of the relevant chemistry and biochemical pathways. Focusing exemplarily on brain and breast cancer, recent advances spanning 2017 to 2023 in conjugated nanocarriers and small molecule drug conjugates were considered, evaluating the chemical and biological aspects in order to improve accessibility to the field and to bridge chemical and biomedical points of view ultimately guiding future research in FR-targeted cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Mohsen Ahmadi
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| | - Christoph A Ritter
- Institute of Pharmacy, Section Clinical Pharmacy, University of Greifswald Greifswald Germany
| | - Thomas von Woedtke
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center Ferdinand-Sauerbruch-Straße 17475 Greifswald Germany
| | - Sander Bekeschus
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
- Clinic and Policlinic for Dermatology and Venereology, Rostock University Medical Center Strempelstr. 13 18057 Rostock Germany
| | - Kristian Wende
- Leibniz Institute for Plasma Science and Technology (INP), Center for Innovation Competence (ZIK) Plasmatis Felix Hausdorff-Str. 2 17489 Greifswald Germany
| |
Collapse
|
13
|
Wang Y, Zhang L, Liu C, Luo Y, Chen D. Peptide-Mediated Nanocarriers for Targeted Drug Delivery: Developments and Strategies. Pharmaceutics 2024; 16:240. [PMID: 38399294 PMCID: PMC10893007 DOI: 10.3390/pharmaceutics16020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Effective drug delivery is essential for cancer treatment. Drug delivery systems, which can be tailored to targeted transport and integrated tumor therapy, are vital in improving the efficiency of cancer treatment. Peptides play a significant role in various biological and physiological functions and offer high design flexibility, excellent biocompatibility, adjustable morphology, and biodegradability, making them promising candidates for drug delivery. This paper reviews peptide-mediated drug delivery systems, focusing on self-assembled peptides and peptide-drug conjugates. It discusses the mechanisms and structural control of self-assembled peptides, the varieties and roles of peptide-drug conjugates, and strategies to augment peptide stability. The review concludes by addressing challenges and future directions.
Collapse
Affiliation(s)
- Yubo Wang
- Medical College, Guangxi University, Da-Xue-Dong Road No. 100, Nanning 530004, China;
| | - Lu Zhang
- School of Life Sciences, Xiamen University, Xiamen 361005, China;
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| | - Chen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| | - Yiming Luo
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, 55 Zhenhai Road, Xiamen 361003, China
- The School of Clinical Medicine, Fujian Medical University, Fuzhou 351002, China
| | - Dengyue Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, China;
| |
Collapse
|
14
|
Tantra T, Singh Y, Patekar R, Kulkarni S, Kumar P, Thareja S. Phosphate Prodrugs: An Approach to Improve the Bioavailability of Clinically Approved Drugs. Curr Med Chem 2024; 31:336-357. [PMID: 36757029 DOI: 10.2174/0929867330666230209094738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 02/10/2023]
Abstract
The phosphate prodrug approach has emerged as a viable option for increasing the bioavailability of a drug candidate with low hydrophilicity and poor cell membrane permeability. When a phosphoric acid moiety is attached to the parent drug, it results in a several-fold elevation in aqueous solubility which helps to achieve desired bioavailability of the pharmaceutically active parental molecule. The neutral phosphate prodrugs have rapid diffusion ability through the plasma membrane as compared to their charged counterpart. The presence of phosphate mono ester breaking alkaline phosphatase (ALP) enzyme throughout the whole human body, is the main consideration behind the development of phosphate prodrug strategy. The popularity of this phosphate prodrug strategy is increasing nowadays due to the fulfillment of different desired pharmacokinetic characteristics required to get pharmaceutical and therapeutic responses without showing any serious adverse drug reactions (ADR). This review article mainly focuses on various phosphate prodrugs synthesized within the last decade to get an improved pharmacological response of the parent moiety along with various preclinical and clinical challenges associated with this approach. Emphasis is also given to the chemical mechanism to release the parent moiety from the prodrug.
Collapse
Affiliation(s)
- Tanmoy Tantra
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Yogesh Singh
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Rohan Patekar
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Swanand Kulkarni
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| |
Collapse
|
15
|
Bisbal Lopez L, Ravazza D, Bocci M, Zana A, Principi L, Dakhel Plaza S, Galbiati A, Gilardoni E, Scheuermann J, Neri D, Pignataro L, Gennari C, Cazzamalli S, Dal Corso A. Ex vivo mass spectrometry-based biodistribution analysis of an antibody-Resiquimod conjugate bearing a protease-cleavable and acid-labile linker. Front Pharmacol 2023; 14:1320524. [PMID: 38125888 PMCID: PMC10731371 DOI: 10.3389/fphar.2023.1320524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Immune-stimulating antibody conjugates (ISACs) equipped with imidazoquinoline (IMD) payloads can stimulate endogenous immune cells to kill cancer cells, ultimately inducing long-lasting anticancer effects. A novel ISAC was designed, featuring the IMD Resiquimod (R848), a tumor-targeting antibody specific for Carbonic Anhydrase IX (CAIX) and the protease-cleavable Val-Cit-PABC linker. In vitro stability analysis showed not only R848 release in the presence of the protease Cathepsin B but also under acidic conditions. The ex vivo mass spectrometry-based biodistribution data confirmed the low stability of the linker-drug connection while highlighting the selective accumulation of the IgG in tumors and its long circulatory half-life.
Collapse
Affiliation(s)
| | | | - Matilde Bocci
- R&D Department, Philochem AG, Otelfingen, Switzerland
| | | | | | | | | | | | - Jörg Scheuermann
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Dario Neri
- R&D Department, Philochem AG, Otelfingen, Switzerland
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
- Philogen S.p.A, Siena, Italy
| | - Luca Pignataro
- Chemistry Department, Università degli Studi di Milano, Milano, Italy
| | - Cesare Gennari
- Chemistry Department, Università degli Studi di Milano, Milano, Italy
| | | | - Alberto Dal Corso
- Chemistry Department, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
16
|
Gonciarz RL, Jiang H, Tram L, Hugelshofer CL, Ekpenyong O, Knemeyer I, Aron AT, Chang CJ, Flygare JA, Collisson EA, Renslo AR. In vivo bioluminescence imaging of labile iron in xenograft models and liver using FeAL-1, an iron-activatable form of D-luciferin. Cell Chem Biol 2023; 30:1468-1477.e6. [PMID: 37820725 PMCID: PMC10841594 DOI: 10.1016/j.chembiol.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 07/21/2023] [Accepted: 09/18/2023] [Indexed: 10/13/2023]
Abstract
Dysregulated iron homeostasis underlies diverse pathologies, from ischemia-reperfusion injury to epithelial-mesenchymal transition and drug-tolerant "persister" cancer cell states. Here, we introduce ferrous iron-activatable luciferin-1 (FeAL-1), a small-molecule probe for bioluminescent imaging of the labile iron pool (LIP) in luciferase-expressing cells and animals. We find that FeAL-1 detects LIP fluctuations in cells after iron supplementation, depletion, or treatment with hepcidin, the master regulator of systemic iron in mammalian physiology. Utilizing FeAL-1 and a dual-luciferase reporter system, we quantify LIP in mouse liver and three different orthotopic pancreatic ductal adenocarcinoma tumors. We observed up to a 10-fold increase in FeAL-1 bioluminescent signal in xenograft tumors as compared to healthy liver, the major organ of iron storage in mammals. Treating mice with hepcidin further elevated hepatic LIP, as predicted. These studies reveal a therapeutic index between tumoral and hepatic LIP and suggest an approach to sensitize tumors toward LIP-activated therapeutics.
Collapse
Affiliation(s)
- Ryan L Gonciarz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Honglin Jiang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Linh Tram
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Cedric L Hugelshofer
- Department of Discovery Chemistry, Merck & Co, Inc., South San Francisco, CA 94080, USA
| | - Oscar Ekpenyong
- ADME & Discovery Toxicology, Merck & Co, Inc., South San Francisco, CA 94080, USA
| | - Ian Knemeyer
- ADME & Discovery Toxicology, Merck & Co, Inc., South San Francisco, CA 94080, USA
| | - Allegra T Aron
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO 80208, USA
| | - Christopher J Chang
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - John A Flygare
- Department of Discovery Chemistry, Merck & Co, Inc., South San Francisco, CA 94080, USA
| | - Eric A Collisson
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
17
|
Mangla P, Vicentini Q, Biscans A. Therapeutic Oligonucleotides: An Outlook on Chemical Strategies to Improve Endosomal Trafficking. Cells 2023; 12:2253. [PMID: 37759475 PMCID: PMC10527716 DOI: 10.3390/cells12182253] [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: 06/21/2023] [Revised: 08/30/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The potential of oligonucleotide therapeutics is undeniable as more than 15 drugs have been approved to treat various diseases in the liver, central nervous system (CNS), and muscles. However, achieving effective delivery of oligonucleotide therapeutics to specific tissues still remains a major challenge, limiting their widespread use. Chemical modifications play a crucial role to overcome biological barriers to enable efficient oligonucleotide delivery to the tissues/cells of interest. They provide oligonucleotide metabolic stability and confer favourable pharmacokinetic/pharmacodynamic properties. This review focuses on the various chemical approaches implicated in mitigating the delivery problem of oligonucleotides and their limitations. It highlights the importance of linkers in designing oligonucleotide conjugates and discusses their potential role in escaping the endosomal barrier, a bottleneck in the development of oligonucleotide therapeutics.
Collapse
Affiliation(s)
- Priyanka Mangla
- Oligonucleotide Discovery, Discovery Sciences Research and Development, AstraZeneca, 431 38 Gothenburg, Sweden; (P.M.); (Q.V.)
| | - Quentin Vicentini
- Oligonucleotide Discovery, Discovery Sciences Research and Development, AstraZeneca, 431 38 Gothenburg, Sweden; (P.M.); (Q.V.)
- Department of Laboratory Medicine, Clinical Research Centre, Karolinska Institute, 141 57 Stockholm, Sweden
| | - Annabelle Biscans
- Oligonucleotide Discovery, Discovery Sciences Research and Development, AstraZeneca, 431 38 Gothenburg, Sweden; (P.M.); (Q.V.)
| |
Collapse
|
18
|
Bugatti K. A Brief Guide to Preparing a Peptide-Drug Conjugate. Chembiochem 2023; 24:e202300254. [PMID: 37288718 DOI: 10.1002/cbic.202300254] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/09/2023]
Abstract
Peptide-drug conjugates (PDCs) have recently emerged as interesting hybrid constructs not only for targeted therapy, but also for the early diagnosis of different pathologies. In most cases, the crucial step in the PDC synthesis is the final conjugation step, where a specific drug is bound to a particular peptide-/peptidomimetic-targeting unit. Thus, this concept paper aims to give a short guide to determining the finest conjugation reaction, by considering in particular the reaction conditions, the stability of the linker and the major pros and cons of each reaction. Based on the recent PDCs reported in literature, the most common and efficient conjugation methods will be systematically presented and compared, generating a short guide to consult while planning the synthesis of a novel peptide-drug conjugate.
Collapse
Affiliation(s)
- Kelly Bugatti
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| |
Collapse
|
19
|
Chiu PF, Chang CK, Huang PS, Lin YY, Lin CS, Yang HY, Hsu LC, Yu LCH, Liang PH. Design, Synthesis, and Evaluation of Glucose Transporter Inhibitor-SN38 Conjugates for Targeting Colorectal Cancer. J Med Chem 2023. [PMID: 37413981 DOI: 10.1021/acs.jmedchem.3c00476] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Irinotecan (1), a prodrug of SN38 (2) approved by the US Food and Drug Administration for treating colorectal cancer, lacks specificity and causes many side effects. To increase the selectivity and therapeutic efficacy of this drug, we designed and synthesized conjugates of SN38 and glucose transporter inhibitors (phlorizin (5) or phloretin (6)), which could be hydrolyzed by glutathione or cathepsin to release SN38 in the tumor microenvironment, as a proof of concept. These conjugates (8, 9, and 10) displayed better antitumor efficacy with lower systemic exposure to SN38 in an orthotopic colorectal cancer mouse model compared with irinotecan at the same dosage. Further, no major adverse effects of the conjugates were observed during treatment. Biodistribution studies showed that conjugate 10 could induce higher concentrations of free SN38 in tumor tissues than irinotecan at the same dosage. Thus, the developed conjugates exhibit potential for treating colorectal cancer.
Collapse
Affiliation(s)
- Pei-Fang Chiu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chun-Kai Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Pin-Shuo Huang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - You-Yu Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chung-Shun Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Hui-Yi Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Pi-Hui Liang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- The Genomics Research Center, Academia Sinica, Taipei 128, Taiwan
| |
Collapse
|
20
|
Zhao H, Yu J, Zhang R, Chen P, Jiang H, Yu W. Doxorubicin prodrug-based nanomedicines for the treatment of cancer. Eur J Med Chem 2023; 258:115612. [PMID: 37441851 DOI: 10.1016/j.ejmech.2023.115612] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023]
Abstract
The chemotherapeutic drug of doxorubicin (DOX) has witnessed widespread applications for treating various cancers. DOX-treated dying cells bear cellular modifications which allow enhanced presentation of tumor antigen and neighboring dendritic cell activation. Furthermore, DOX also facilitate the immune-mediated clearance of tumor cells. However, disadvantages such as severe off-target toxicity, and prominent hydrophobicity have resulted in unsatisfactory clinical therapeutic outcomes. The effective delivery of DOX drug molecules is still challenging despite the rapid advances in nanotechnology and biomaterials. Huge progress has been witnessed in DOX nanoprodrugs owing to their brilliant benefits such as tumor stimuli-responsive drug release capacity, high drug loading efficiency and so on. This review summarized recent progresses of DOX prodrug-based nanomedicines to provide deep insights into future development and inspire researchers to explore DOX nanoprodrugs with real clinical applications.
Collapse
Affiliation(s)
- Haibo Zhao
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Renshuai Zhang
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China
| | - Pengwei Chen
- Hainan Key Laboratory for Research and Development of Natural Product from Li Folk Medicine, Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, China
| | - Hongfei Jiang
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, China.
| | - Wanpeng Yu
- Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
21
|
Gonciarz RL, Sakhamuri S, Hooshdaran N, Kumar G, Kim H, Evans MJ, Renslo AR. Elevated labile iron in castration-resistant prostate cancer is targetable with ferrous iron-activatable antiandrogen therapy. Eur J Med Chem 2023; 249:115110. [PMID: 36708680 PMCID: PMC10210592 DOI: 10.1016/j.ejmech.2023.115110] [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: 08/26/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Clinical responses to second generation androgen signaling inhibitors (e.g., enzalutamide) in metastatic castration-resistant prostate cancer (mCRPC) are variable and transient, and are associated with dose limiting toxicities, including rare but severe CNS effects. We hypothesized that changes to iron metabolism coincident with more advanced disease might be leveraged for tumor-selective delivery of antiandrogen therapy. Using the recently described chemical probes SiRhoNox and 18F-TRX in mCRPC models, we found elevated Fe2+ to be a common feature of mCRPC in vitro and in vivo. We next synthesized ferrous-iron activatable drug conjugates of second and third-generation antiandrogens and found these conjugates possessed comparable or enhanced antiproliferative activity across mCRPC cell line models. Mouse pharmacokinetic studies showed that these prototype antiandrogen conjugates are stable in vivo and limited exposure to conjugate or free antiandrogen in the brain. Our results reveal elevated Fe2+ to be a feature of mCRPC that might be leveraged to improve the tolerability and efficacy of antiandrogen therapy.
Collapse
Affiliation(s)
- Ryan L Gonciarz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, 94158, United States
| | - Sasank Sakhamuri
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94158, United States
| | - Nima Hooshdaran
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94158, United States
| | - Garima Kumar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94158, United States
| | - Hyunjung Kim
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94158, United States
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94158, United States; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94158, United States.
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, 94158, United States; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94158, United States.
| |
Collapse
|
22
|
Paulus J, Nachtigall B, Meyer P, Sewald N. RGD Peptidomimetic MMAE-Conjugate Addressing Integrin αVβ3-Expressing Cells with High Targeting Index. Chemistry 2023; 29:e202203476. [PMID: 36454662 DOI: 10.1002/chem.202203476] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/03/2022]
Abstract
Small molecule-drug conjugates (SMDCs) mimicking the RGD sequence (-Arg-Gly-Asp-) with a non-peptide moiety require a pharmacophore-independent attachment site. A library of 36 sulfonamide-modified RGD mimetics with nM to pM affinity for integrin αV β3 was synthesized and analysed via DAD mapping. The best structure of the conjugable RGD mimetic was used and a linker was attached to an aromatic ring by Negishi cross-coupling. The product retained high affinity and selectivity for integrin αV β3 . The conjugable RGD mimetic was then attached to an enzymatically cleavable GKGEVA linker equipped with a self-immolative PABC and the antimitotic drug monomethyl auristatin E (MMAE). The resulting SMDC preferred binding to integrin αV β3 over α5 β1 in a ratio of 1 : 4519 (ELISA) and showed selectivity for αV β3 -positive WM115 cells over αV β3 -negative M21-L cells in the in vitro cell adhesion assay as well as in cell viability assays with a targeting index of 134 (M21-L/WM115).
Collapse
Affiliation(s)
- Jannik Paulus
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Beate Nachtigall
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Peter Meyer
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| |
Collapse
|
23
|
Bessaguet A, Blancart‐Remaury Q, Poinot P, Opalinski I, Papot S. Stimuli-Responsive Catenane-Based Catalysts. Angew Chem Int Ed Engl 2023; 62:e202216787. [PMID: 36478644 PMCID: PMC10107136 DOI: 10.1002/anie.202216787] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Rotaxanes and molecular knots exhibit particular properties resulting from the presence of a mechanical bond within their structure that maintains the molecular components interlocked in a permanent manner. On the other hand, the disassembly of the interlocked architecture through the breakdown of the mechanical bond can activate properties which are masked in the parent compound. Herein, we present the development of stimuli-responsive CuI -complexed [2]catenanes as OFF/ON catalysts for the copper-catalyzed alkyne-azide cycloaddition (CuAAC) reaction. The encapsulation of the CuI ion inside the [2]catenanes inhibits its ability to catalyze the formation of triazoles. In contrast, the controlled opening of the two macrocycles induces the breaking of the mechanical bond, thereby restoring the catalytic activity of the CuI ion for the CuAAC reaction. Such OFF/ON catalysts can be involved in signal amplification processes with various potential applications.
Collapse
Affiliation(s)
- Adrien Bessaguet
- University of PoitiersUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)4 rue Michel-Brunet, TSA 5110686073Poitiers cedex 9France
| | - Quentin Blancart‐Remaury
- University of PoitiersUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)4 rue Michel-Brunet, TSA 5110686073Poitiers cedex 9France
| | - Pauline Poinot
- University of PoitiersUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)4 rue Michel-Brunet, TSA 5110686073Poitiers cedex 9France
| | - Isabelle Opalinski
- University of PoitiersUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)4 rue Michel-Brunet, TSA 5110686073Poitiers cedex 9France
| | - Sébastien Papot
- University of PoitiersUMR CNRS 7285Institut de Chimie des Milieux et Matériaux de Poitiers (IC2MP)4 rue Michel-Brunet, TSA 5110686073Poitiers cedex 9France
| |
Collapse
|
24
|
Ribéraud M, Porte K, Chevalier A, Madegard L, Rachet A, Delaunay-Moisan A, Vinchon F, Thuéry P, Chiappetta G, Champagne PA, Pieters G, Audisio D, Taran F. Fast and Bioorthogonal Release of Isocyanates in Living Cells from Iminosydnones and Cycloalkynes. J Am Chem Soc 2023; 145:2219-2229. [PMID: 36656821 DOI: 10.1021/jacs.2c09865] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Bioorthogonal click-and-release reactions are powerful tools for chemical biology, allowing, for example, the selective release of drugs in biological media, including inside animals. Here, we developed two new families of iminosydnone mesoionic reactants that allow a bioorthogonal release of electrophilic species under physiological conditions. Their synthesis and reactivities as dipoles in cycloaddition reactions with strained alkynes have been studied in detail. Whereas the impact of the pH on the reaction kinetics was demonstrated experimentally, theoretical calculations suggest that the newly designed dipoles display reduced resonance stabilization energies compared to previously described iminosydnones, explaining their higher reactivity. These mesoionic compounds react smoothly with cycloalkynes under physiological, copper-free reaction conditions to form a click pyrazole product together with a released alkyl- or aryl-isocyanate. With rate constants up to 1000 M-1 s-1, this click-and-release reaction is among the fastest described to date and represents the first bioorthogonal process allowing the release of isocyanate electrophiles inside living cells, offering interesting perspectives in chemical biology.
Collapse
Affiliation(s)
- Maxime Ribéraud
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France
| | - Karine Porte
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France
| | - Arnaud Chevalier
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198 Gif-sur-Yvette, France
| | - Léa Madegard
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France
| | - Aurélie Rachet
- Université Paris Saclay, CEA, Institut de Biologie Intégrative de la Cellule (I2BC), 91191 Gif-sur-Yvette, France
| | - Agnès Delaunay-Moisan
- Université Paris Saclay, CEA, Institut de Biologie Intégrative de la Cellule (I2BC), 91191 Gif-sur-Yvette, France
| | - Florian Vinchon
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France
| | - Pierre Thuéry
- Université Paris-Saclay, CEA, CNRS, NIMBE, 91191 Gif-sur-Yvette, France
| | - Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005 Paris, France
| | - Pier Alexandre Champagne
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, New Jersey 07102, United States
| | - Grégory Pieters
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France
| | - Davide Audisio
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France
| | - Frédéric Taran
- Département Médicaments et Technologies pour la Santé (DMTS), SCBM, Université Paris Saclay, CEA, INRAE, 91191 Gif-sur-Yvette, France
| |
Collapse
|
25
|
Yang Y, Wang S, Ma P, Jiang Y, Cheng K, Yu Y, Jiang N, Miao H, Tang Q, Liu F, Zha Y, Li N. Drug conjugate-based anticancer therapy - Current status and perspectives. Cancer Lett 2023; 552:215969. [PMID: 36279982 DOI: 10.1016/j.canlet.2022.215969] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
Drug conjugates are conjugates comprising a tumor-homing carrier tethered to a cytotoxic agent via a linker that are designed to deliver an ultra-toxic payload directly to the target cancer cells. This strategy has been successfully used to increase the therapeutic efficacy of cytotoxic agents and reduce their toxic side effects. Drug conjugates are being developed worldwide, with the potential to revolutionize current cancer treatment strategies. Antibody-drug conjugates (ADCs) have developed rapidly, and 14 of them have received market approval since the first approval event by the Food and Drug Administration in 2000. However, there are some limitations in the use of antibodies as carriers. Other classes of drug conjugates are emerging, such as targeted drugs conjugated with peptides (peptide-drug conjugates, PDCs) and polymers (polymer-drug conjugates, PolyDCs) with the remaining constructs similar to those of ADCs. These novel drug conjugates are gaining attention because they overcome the limitations of ADCs. This review summarizes the current state and advancements in knowledge regarding the design, constructs, and clinical efficacy of different drug conjugates.
Collapse
Affiliation(s)
- Yuqi Yang
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Shuhang Wang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peiwen Ma
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yale Jiang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Yue Yu
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ning Jiang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Huilei Miao
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qiyu Tang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Funan Liu
- First Affiliated Hospital of China Medical University, Shenyang, 110002, China
| | - Yan Zha
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| | - Ning Li
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
26
|
Yamazaki S, Matsuda Y. Tag‐Free Enzymatic Modification for Antibody−Drug Conjugate Production. ChemistrySelect 2022. [DOI: 10.1002/slct.202203753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Yutaka Matsuda
- Ajinomoto Bio-Pharma Services 11040 Roselle Street San Diego CA 92121 United States
| |
Collapse
|
27
|
Pignataro L, Piarulli U. A Long Journey through Organic Chemistry – Celebrating Cesare Gennari's 70th Birthday. European J Org Chem 2022. [DOI: 10.1002/ejoc.202201160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Luca Pignataro
- Dipartimento di Chimica Università degli Studi di Milano Via C. Golgi, 19 20133 Milano Italy
| | - Umberto Piarulli
- Università degli Studi dell'Insubria Dipartimento di Scienza e Alta Tecnologia Via Valleggio, 11 I-22100 Como Italy
| |
Collapse
|
28
|
Keppel P, Sohr B, Kuba W, Goldeck M, Skrinjar P, Carlson JCT, Mikula H. Tetrazine-Triggered Bioorthogonal Cleavage of trans-Cyclooctene-Caged Phenols Using a Minimal Self-Immolative Linker Strategy. Chembiochem 2022; 23:e202200363. [PMID: 35921044 PMCID: PMC9804162 DOI: 10.1002/cbic.202200363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/02/2022] [Indexed: 01/05/2023]
Abstract
Bond-cleavage reactions triggered by bioorthogonal tetrazine ligation have emerged as strategies to chemically control the function of (bio)molecules and achieve activation of prodrugs in living systems. While most of these approaches make use of caged amines, current methods for the release of phenols are limited by unfavorable reaction kinetics or insufficient stability of the Tz-responsive reactants. To address this issue, we have implemented a self-immolative linker that enables the connection of cleavable trans-cyclooctenes (TCO) and phenols via carbamate linkages. Based on detailed investigation of the reaction mechanism with several Tz, revealing up to 96 % elimination after 2 hours, we have developed a TCO-caged prodrug with 750-fold reduced cytotoxicity compared to the parent drug and achieved in situ activation upon Tz/TCO click-to-release.
Collapse
Affiliation(s)
- Patrick Keppel
- Institute of Applied Synthetic ChemistryTU Wien1060ViennaAustria
| | - Barbara Sohr
- Institute of Applied Synthetic ChemistryTU Wien1060ViennaAustria
| | - Walter Kuba
- Institute of Applied Synthetic ChemistryTU Wien1060ViennaAustria
| | - Marion Goldeck
- Institute of Applied Synthetic ChemistryTU Wien1060ViennaAustria
- Center for Anatomy and Cell BiologyMedical University of Vienna1090ViennaAustria
| | - Philipp Skrinjar
- Institute of Applied Synthetic ChemistryTU Wien1060ViennaAustria
| | - Jonathan C. T. Carlson
- Center for Systems Biology & Department of MedicineMassachusetts General HospitalHarvard Medical SchoolBoston, MA02114USA
| | - Hannes Mikula
- Institute of Applied Synthetic ChemistryTU Wien1060ViennaAustria
| |
Collapse
|
29
|
Glycoconjugation of Quinoline Derivatives Using the C-6 Position in Sugars as a Strategy for Improving the Selectivity and Cytotoxicity of Functionalized Compounds. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27206918. [PMID: 36296513 PMCID: PMC9607644 DOI: 10.3390/molecules27206918] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/19/2022]
Abstract
Based on the Warburg effect and the increased demand for glucose by tumor cells, a targeted drug delivery strategy was developed. A series of new glycoconjugates with increased ability to interact with GLUT transporters, responsible for the transport of sugars to cancer cells, were synthesized. Glycoconjugation was performed using the C-6 position in the sugar unit, as the least involved in the formation of hydrogen bonds with various aminoacids residues of the transporter. The carbohydrate moiety was connected with the 8-hydroxyquinoline scaffold via a 1,2,3-triazole linker. For the obtained compounds, several in vitro biological tests were performed using HCT-116 and MCF-7 cancer cells as well as NHDF-Neo healthy cells. The highest cytotoxicity of both cancer cell lines in the MTT test was noted for glycoconjugates in which the triazole-quinoline was attached through the triazole nitrogen atom to the d-glucose unit directly to the carbon at the C-6 position. These compounds were more selective than the analogous glycoconjugates formed by the C-1 anomeric position of d-glucose. Experiments with an EDG inhibitor have shown that GLUTs can be involved in the transport of glycoconjugates. The results of apoptosis and cell cycle analyses by flow cytometry confirmed that the new type of glycoconjugates shows pro-apoptotic properties, without significantly affecting changes in the distribution of the cell cycle. Moreover, glycoconjugates were able to decrease the clonogenic potential of cancer cells, inhibit the migration capacity of cells and intercalate with DNA.
Collapse
|
30
|
Migliorini F, Cini E, Dreassi E, Finetti F, Ievoli G, Macrì G, Petricci E, Rango E, Trabalzini L, Taddei M. A pH-responsive crosslinker platform for antibody-drug conjugate (ADC) targeting delivery. Chem Commun (Camb) 2022; 58:10532-10535. [PMID: 36043993 DOI: 10.1039/d2cc03052g] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a new 1-6 self-immolative, traceless crosslinker derived from the natural product gallic acid. The linker acts through a pH-dependent mechanism for drug release. This 5-(hydroxymethyl)pyrogallol orthoester derivative (HMPO) was stable for 24 hours at pH values of 7.4 and 6.6 and in plasma, releasing molecules bound to the hydroxymethyl moiety under acid-dependent stimuli at pH 5.5. The linker was non-toxic and was used for the conjugation of Doxorubicin (Doxo) or Combretastatin A4 with Cetuximab. The ADCs formed showed their pH responsivity reducing cell viability of A431 and A549 cancer cells better than Cetuximab alone.
Collapse
Affiliation(s)
- Francesca Migliorini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Elena Cini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Elena Dreassi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Federica Finetti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Giovanni Ievoli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Giulia Macrì
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Elena Petricci
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Enrico Rango
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Lorenza Trabalzini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| | - Maurizio Taddei
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A.Moro 2, 53100 Siena, Italy.
| |
Collapse
|
31
|
Han S, Lim KS, Blackburn BJ, Yun J, Putnam CW, Bull DA, Won YW. The Potential of Topoisomerase Inhibitor-Based Antibody–Drug Conjugates. Pharmaceutics 2022; 14:pharmaceutics14081707. [PMID: 36015333 PMCID: PMC9413092 DOI: 10.3390/pharmaceutics14081707] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 12/17/2022] Open
Abstract
DNA topoisomerases are essential enzymes that stabilize DNA supercoiling and resolve entanglements. Topoisomerase inhibitors have been widely used as anti-cancer drugs for the past 20 years. Due to their selectivity as topoisomerase I (TOP1) inhibitors that trap TOP1 cleavage complexes, camptothecin and its derivatives are promising anti-cancer drugs. To increase accumulation of TOP1 inhibitors in cancer cells through the targeting of tumors, TOP1 inhibitor antibody–drug conjugates (TOP1-ADC) have been developed and marketed. Some TOP1-ADCs have shown enhanced therapeutic efficacy compared to prototypical anti-cancer ADCs, such as T-DM1. Here, we review various types of camptothecin-based TOP1 inhibitors and recent developments in TOP1-ADCs. We then propose key points for the design and construction of TOP1-ADCs. Finally, we discuss promising combinatorial strategies, including newly developed approaches to maximizing the therapeutic potential of TOP1-ADCs.
Collapse
Affiliation(s)
- Seungmin Han
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - Kwang Suk Lim
- Department of Biotechnology and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon 24341, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon 24341, Korea
| | - Brody J. Blackburn
- Department of Medical Pharmacology, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - Jina Yun
- Division of Hematology-Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Korea
| | - Charles W. Putnam
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - David A. Bull
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - Young-Wook Won
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
- Correspondence:
| |
Collapse
|
32
|
Dal Corso A, Frigoli M, Prevosti M, Mason M, Bucci R, Belvisi L, Pignataro L, Gennari C. Advanced Pyrrolidine-Carbamate Self-Immolative Spacer with Tertiary Amine Handle Induces Superfast Cyclative Drug Release. ChemMedChem 2022; 17:e202200279. [PMID: 35620983 PMCID: PMC9544318 DOI: 10.1002/cmdc.202200279] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Indexed: 11/07/2022]
Abstract
Amine-carbamate self-immolative (SI) spacers represent practical and versatile tools in targeted prodrugs, but their slow degradation mechanism limits drug activation at the site of disease. We engineered a pyrrolidine-carbamate SI spacer with a tertiary amine handle which strongly accelerates the spacer cyclization to give a bicyclic urea and the free hydroxy groups of either cytotoxic (Camptothecin) or immunostimulatory (Resiquimod) drugs. In silico conformational analysis and pKa calculations suggest a plausible mechanism for the superior efficacy of the advanced SI spacer compared to state-of-art analogues.
Collapse
Affiliation(s)
- Alberto Dal Corso
- Università degli Studi di MilanoDipartimento di Chimicavia C. Golgi, 1920133MilanItaly
| | - Margaux Frigoli
- Università degli Studi di MilanoDipartimento di Chimicavia C. Golgi, 1920133MilanItaly
| | - Martina Prevosti
- Università degli Studi di MilanoDipartimento di Chimicavia C. Golgi, 1920133MilanItaly
| | - Mattia Mason
- Università degli Studi di MilanoDipartimento di Chimicavia C. Golgi, 1920133MilanItaly
| | - Raffaella Bucci
- Università degli Studi di MilanoDipartimento di Scienze Farmaceutichevia G. Venezian 2120133MilanItaly
| | - Laura Belvisi
- Università degli Studi di MilanoDipartimento di Chimicavia C. Golgi, 1920133MilanItaly
| | - Luca Pignataro
- Università degli Studi di MilanoDipartimento di Chimicavia C. Golgi, 1920133MilanItaly
| | - Cesare Gennari
- Università degli Studi di MilanoDipartimento di Chimicavia C. Golgi, 1920133MilanItaly
| |
Collapse
|
33
|
Evans N, Grygorash R, Williams P, Kyle A, Kantner T, Pathak R, Sheng X, Simoes F, Makwana H, Resende R, de Juan E, Jenkins A, Morris D, Michelet A, Jewitt F, Rudge F, Camper N, Manin A, McDowell W, Pabst M, Godwin A, Frigerio M, Bird M. Incorporation of Hydrophilic Macrocycles Into Drug-Linker Reagents Produces Antibody-Drug Conjugates With Enhanced in vivo Performance. Front Pharmacol 2022; 13:764540. [PMID: 35784686 PMCID: PMC9247464 DOI: 10.3389/fphar.2022.764540] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Antibody-drug conjugates (ADCs) have begun to fulfil their promise as targeted cancer therapeutics with ten clinical approvals to date. As the field matures, much attention has focused upon the key factors required to produce safe and efficacious ADCs. Recently the role that linker-payload reagent design has on the properties of ADCs has been highlighted as an important consideration for developers. We have investigated the effect of incorporating hydrophilic macrocycles into reagent structures on the in vitro and in vivo behavior of ADCs. Bis-sulfone based disulfide rebridging reagents bearing Val-Cit-PABC-MMAE linker-payloads were synthesized with a panel of cyclodextrins and crown ethers integrated into their structures via a glutamic acid branching point. Brentuximab was selected as a model antibody and ten ADCs with a drug-to-antibody ratio (DAR) of 4 were prepared for biological evaluation. In vitro, the ADCs prepared showed broadly similar potency (range: 16–34 pM) and were comparable to Adcetris® (16 pM). In vivo, the cyclodextrin containing ADCs showed greater efficacy than Adcetris® and the most efficacious variant (incorporating a 3′-amino-α-cyclodextrin component) matched a 24-unit poly(ethylene glycol) (PEG) containing comparator. The ADCs bearing crown ethers also displayed enhanced in vivo efficacy compared to Adcetris®, the most active variant (containing a 1-aza-42-crown-14 macrocycle) was superior to an analogous ADC with a larger 24-unit PEG chain. In summary, we have demonstrated that hydrophilic macrocycles can be effectively incorporated into ADC reagent design and offer the potential for enhanced alternatives to established drug-linker architectures.
Collapse
|
34
|
Johan AN, Li Y. Development of Photoremovable Linkers as a Novel Strategy to Improve the Pharmacokinetics of Drug Conjugates and Their Potential Application in Antibody-Drug Conjugates for Cancer Therapy. Pharmaceuticals (Basel) 2022; 15:655. [PMID: 35745573 PMCID: PMC9230074 DOI: 10.3390/ph15060655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/19/2022] [Accepted: 05/22/2022] [Indexed: 02/04/2023] Open
Abstract
Although there have been extensive research and progress on the discovery of anticancer drug over the years, the application of these drugs as stand-alone therapy has been limited by their off-target toxicities, poor pharmacokinetic properties, and low therapeutic index. Targeted drug delivery, especially drug conjugate, has been recognized as a technology that can bring forth a new generation of therapeutics with improved efficacy and reduced side effects for cancer treatment. The linker in a drug conjugate is of essential importance because it impacts the circulation time of the conjugate and the release of the drug for full activity at the target site. Recently, the light-triggered linker has attracted a lot of attention due to its spatiotemporal controllability and attractive prospects of improving the overall pharmacokinetics of the conjugate. In this paper, the latest developments of UV- and IR-triggered linkers and their application and potential in drug conjugate development are reviewed. Some of the most-well-researched photoresponsive structural moieties, such as UV-triggered coumarin, ortho-nitrobenzyl group (ONB), thioacetal ortho-nitrobenzaldehyde (TNB), photocaged C40-oxidized abasic site (PC4AP), and IR-triggered cyanine and BODIPY, are included for discussion. These photoremovable linkers show better physical and chemical stabilities and can undergo rapid cleavage upon irradiation. Very importantly, the drug conjugates containing these linkers exhibit reduced off-target toxicity and overall better pharmacokinetic properties. The progress on photoactive antibody-drug conjugates, such as antibody-drug conjugates (ADC) and antibody-photoabsorber conjugate (APC), as precision medicine in clinical cancer treatment is highlighted.
Collapse
Affiliation(s)
| | - Yi Li
- Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China
| |
Collapse
|
35
|
Javia A, Vanza J, Bardoliwala D, Ghosh S, Misra A, Patel M, Thakkar H. Polymer-drug conjugates: Design principles, emerging synthetic strategies and clinical overview. Int J Pharm 2022; 623:121863. [PMID: 35643347 DOI: 10.1016/j.ijpharm.2022.121863] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/06/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
Adagen, an enzyme replacement treatment for adenosine deaminase deficiency, was the first protein-polymer conjugate to be approved in early 1990s. Post this regulatory approval, numerous polymeric drugs and polymeric nanoparticles have entered the market as advanced or next-generation polymer-based therapeutics, while many others have currently been tested clinically. The polymer conjugation to therapeutic moiety offers several advantages, like enhanced solubilization of drug, controlled release, reduced immunogenicity, and prolonged circulation. The present review intends to highlight considerations in the design of therapeutically effective polymer-drug conjugates (PDCs), including the choice of linker chemistry. The potential synthetic strategies to formulate PDCs have been discussed along with recent advancements in the different types of PDCs, i.e., polymer-small molecular weight drug conjugates, polymer-protein conjugates, and stimuli-responsive PDCs, which are under clinical/preclinical investigation. Current impediments and regulatory hurdles hindering the clinical translation of PDC into effective therapeutic regimens for the amelioration of disease conditions have been addressed.
Collapse
Affiliation(s)
- Ankit Javia
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Jigar Vanza
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat-388421, India
| | - Denish Bardoliwala
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India; Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKM's NMIMS, Shirpur, Maharashtra-425405, Indi
| | - Mrunali Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, Changa, Gujarat-388421, India
| | - Hetal Thakkar
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat-390001, India.
| |
Collapse
|
36
|
Collyer SE, Stack GD, Walsh JJ. Selective delivery of clinically approved tubulin binding agents through covalent conjugation to an active targeting moiety. Curr Med Chem 2022; 29:5179-5211. [DOI: 10.2174/0929867329666220401105929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
The efficacy and tolerability of tubulin binding agents are hampered by their low specificity for cancer cells, like most clinically used anticancer agents. To improve specificity, tubulin binding agents have been covalently conjugated to agents which target cancer cells to give actively targeted drug conjugates. These conjugates are designed to increase uptake of the drug by cancer cells, while having limited uptake by normal cells thereby improving efficacy and tolerability.
Approaches used include attachment to small molecules, polysaccharides, peptides, proteins and antibodies that exploit the overexpression of receptors for these substances. Antibody targeted strategies have been the most successful to date with six such examples having gained clinical approval. Many other conjugate types, especially those targeting the folate receptor, have shown promising efficacy and toxicity profiles in pre-clinical models and in early-stage clinical studies. Presented herein is a discussion of the success or otherwise of the recent strategies used to form these actively targeted conjugates.
Collapse
Affiliation(s)
- Samuel E. Collyer
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - Gary D. Stack
- Department of Nursing and Healthcare, Technological University of the Shannon: Midlands Midwest, Athlone, Ireland
| | - John J. Walsh
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Kang J, Mun SK, Choi EJ, Kim JJ, Yee ST, Chang DJ. A preliminary study for the development of cleavable linkers using activatable fluorescent probes targeting leucine aminopeptidase. Analyst 2022; 147:5386-5394. [DOI: 10.1039/d2an01145j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The Kinetic and ex vivo stability studies of LAP-responsive fluorescent probes demonstrated that the stability and intrinsic drug releasing activity of the cleavalbe linker can be evaluated by a peptidase-responsive activatable fluorescent probe.
Collapse
Affiliation(s)
- Julie Kang
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Korea
| | - Seul-Ki Mun
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Korea
| | - Eu-Jin Choi
- Department of Biology, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Korea
| | - Jong-Jin Kim
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61186, Korea
| | - Sung-Tae Yee
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Korea
| | - Dong-Jo Chang
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, 255 Jungang-ro, Suncheon 57922, Korea
| |
Collapse
|
38
|
Sweet-Jones J, Ahmad M, Martin AC. Antibody markup language (AbML) - a notation language for antibody-based drug formats and software for creating and rendering AbML (abYdraw). MAbs 2022; 14:2101183. [PMID: 35838549 PMCID: PMC9291709 DOI: 10.1080/19420862.2022.2101183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/08/2022] [Indexed: 01/07/2023] Open
Abstract
As interest in antibody-based drug development continues to increase, the biopharmaceutical industry has begun to focus on complex multi-specific antibodies (MsAbs) as an up-and-coming class of biologic that differ from natural monoclonal antibodies through their ability to bind to more than one type of antigen. As techniques to generate such molecules have diversified, so have their formats and the need for standard notation. Previous efforts to develop a notation language for macromolecule drugs have been insufficient, or too complex, for MsAbs. Here, we present Antibody Markup Language (AbML), a new notation language specifically for antibody formats that overcomes the limitations of existing languages and can annotate all current antibody formats, including fusions, fragments, standard antibodies and MsAbs, as well as all currently conceivable future formats. AbML V1.1 also provides explicit support for T-cell receptor domains. To assist users of this language we have also developed a tool, abYdraw, that can draw antibody schematics from AbML strings or generate an AbML string from a drawn antibody schematic. AbML has the potential to become a standardized notation for describing new MsAb formats entering clinical trials.Abbreviations: AbML: Antibody Markup Language; ADC: Antibody-drug conjugate; CAS: Chemical Abstracts Service; CH: Constant heavy; CL: Constant light; Fv: Variable fragment; HELM: Hierarchical Editing Language for Macromolecules; HSA: Human serum albumin; INN: International Nonproprietary Names; KIH: Knobs-into-holes; mAbs: Monoclonal antibodies; MsAb: Multi-specific antibody; WHO: World Health Organization; PEG: Poly-ethylene glycol; scFv: Single-chain variable fragment; SMILES: Simplified Molecular-Input Line-Entry System; VH: Variable heavy; VHH: Single-domain (Camelid) variable heavy; VL: Variable light.
Collapse
Affiliation(s)
- James Sweet-Jones
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Maham Ahmad
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Andrew C.R. Martin
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| |
Collapse
|
39
|
Gavriel A, Sambrook M, Russell AT, Hayes W. Recent advances in self-immolative linkers and their applications in polymeric reporting systems. Polym Chem 2022. [DOI: 10.1039/d2py00414c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Interest in self-immolative chemistry has grown over the past decade with more research groups harnessing the versatility to control the release of a compound from a larger chemical entity, given...
Collapse
|
40
|
Usama SM, Marker SC, Caldwell DR, Patel NL, Feng Y, Kalen JD, St Croix B, Schnermann MJ. Targeted Fluorogenic Cyanine Carbamates Enable In Vivo Analysis of Antibody-Drug Conjugate Linker Chemistry. J Am Chem Soc 2021; 143:21667-21675. [PMID: 34928588 DOI: 10.1021/jacs.1c10482] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibody-drug conjugates (ADCs) are a rapidly emerging therapeutic platform. The chemical linker between the antibody and the drug payload plays an essential role in the efficacy and tolerability of these agents. New methods that quantitatively assess the cleavage efficiency in complex tissue settings could provide valuable insights into the ADC design process. Here we report the development of a near-infrared (NIR) optical imaging approach that measures the site and extent of linker cleavage in mouse models. This approach is enabled by a superior variant of our recently devised cyanine carbamate (CyBam) platform. We identify a novel tertiary amine-containing norcyanine, the product of CyBam cleavage, that exhibits a dramatically increased cellular signal due to an improved cellular permeability and lysosomal accumulation. The resulting cyanine lysosome-targeting carbamates (CyLBams) are ∼50× brighter in cells, and we find this strategy is essential for high-contrast in vivo targeted imaging. Finally, we compare a panel of several common ADC linkers across two antibodies and tumor models. These studies indicate that cathepsin-cleavable linkers provide dramatically higher tumor activation relative to hindered or nonhindered disulfides, an observation that is only apparent with in vivo imaging. This strategy enables quantitative comparisons of cleavable linker chemistries in complex tissue settings with implications across the drug delivery landscape.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Sierra C Marker
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Donald R Caldwell
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Nimit L Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
41
|
Huvelle S, Le Saux T, Jullien L, Schmidt F. A double-triggered self-immolative spacer for increased selectivity of molecular release. Org Biomol Chem 2021; 20:240-246. [PMID: 34897358 DOI: 10.1039/d1ob02124a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A self-immolative spacer based on dissymmetrical N,N'-bis-carbamate aniline is introduced to liberate a substrate from a precursor after dual activation. The proof of principle of its exclusive selectivity for substrate liberation has been conducted on a profluorophore.
Collapse
Affiliation(s)
- Steve Huvelle
- i-CleHS, UMR 8060, Chimie ParisTech - PSL, Paris, Île-de-France, France
| | - Thomas Le Saux
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005 Paris, France.
| | - Ludovic Jullien
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005 Paris, France.
| | - Frédéric Schmidt
- Institut Curie, PSL University, CNRS UMR 3666 - INSERM U1143, 26 rue d'Ulm, 75248 PARIS CEDEX 05, FRANCE.
| |
Collapse
|
42
|
Li J, Li X, Liu P. Acid-triggered degradable diblock poly(doxorubicin)-polyethylene glycol polyprodrug with doxorubicin as structural unit for tumor intracellular delivery. Int J Pharm 2021; 609:121142. [PMID: 34600057 DOI: 10.1016/j.ijpharm.2021.121142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/30/2021] [Accepted: 09/25/2021] [Indexed: 10/20/2022]
Abstract
Polyprodrugs, in which drug was used as the structural unit by linking with each other via the dynamic covalent bonds in the main chain, are expected to endow excellent drug delivery performance. Here, acid-triggered degradable diblock polyprodrug, poly(doxorubicin)-polyethylene glycol (PDOX-PEG), was designed with DOX as structural unit alternately linked with acid-labile hydrazone and maleic amide groups, by the polycondensation of DOX-based dimers (D-DOXADH or D-DOXMAH) with PEGylated dimer (DOX-ADH-DOX-PEG) as end capping agent. The optimized PDOX-PEG, which was synthesized with D-DOXADH and the PEGylated dimer at a feeding ratio of 10%, possessed a high Mn of 3.1 × 104 g/mol with a high DOX content of 75.42%. It could easily self-assemble into near spherical nanoparticles with average hydrodynamic diameter of 135 nm. They showed excellent pH-triggered sustained drug release owing to the acid-triggered degradation of the polyprodrug block in the tumor intracellular microenvironment, with low premature drug leakage of 4.39 % within 60 h. The MTT results indicated the enhanced antitumor efficacy of the proposed PDOX-PEG nanoparticles than free DOX. The results demonstrated the promising potential of the proposed acid-triggered degradable diblock PDOX-PEG polyprodrug for tumor treatment.
Collapse
Affiliation(s)
- Jiagen Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
43
|
Raabe M, Heck AJ, Führer S, Schauenburg D, Pieszka M, Wang T, Zegota MM, Nuhn L, Ng DYW, Kuan SL, Weil T. Assembly of pH-Responsive Antibody-Drug-Inspired Conjugates. Macromol Biosci 2021; 22:e2100299. [PMID: 34791790 DOI: 10.1002/mabi.202100299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/11/2021] [Indexed: 01/12/2023]
Abstract
With the advent of chemical strategies that allow the design of smart bioconjugates, peptide- and protein-drug conjugates are emerging as highly efficient therapeutics to overcome limitations of conventional treatment, as exemplified by antibody-drug conjugates (ADCs). While targeting peptides serve similar roles as antibodies to recognize overexpressed receptors on diseased cell surfaces, peptide-drug conjugates suffer from poor stability and bioavailability due to their low molecular weights. Through a combination of a supramolecular protein-based assembly platform and a pH-responsive linker, the authors devise herein the convenient assembly of a trivalent protein-drug conjugate. The conjugate should ideally possess distinct features of ADCs such as 1) recognition sites that recognize cell receptor and are arranged on 2) distinct locations on a high molecular weight protein scaffold, 3) a stimuli-responsive linker, as well as 4) an attached payload such as a drug molecule. These AD-like conjugates target cancer cells that overexpress somatostatin receptors, can enable controlled release in the microenvironment of cancer cells through a new pH-responsive biotin linker, and exhibit stability in biological media.
Collapse
Affiliation(s)
- Marco Raabe
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany.,Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany.,Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Astrid Johanna Heck
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
| | - Siska Führer
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
| | - Dominik Schauenburg
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
| | - Michaela Pieszka
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany.,Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany
| | - Tao Wang
- Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany.,Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, 600213, P. R. China
| | - Maksymilian Marek Zegota
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany.,Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany
| | - Lutz Nuhn
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
| | - David Y W Ng
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany
| | - Seah Ling Kuan
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany.,Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany
| | - Tanja Weil
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz, 55128, Germany.,Institute of Inorganic Chemistry I, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany
| |
Collapse
|
44
|
Baiula M, Cirillo M, Martelli G, Giraldi V, Gasparini E, Anelli AC, Spampinato SM, Giacomini D. Selective Integrin Ligands Promote Cell Internalization of the Antineoplastic Agent Fluorouracil. ACS Pharmacol Transl Sci 2021; 4:1528-1542. [PMID: 34661072 PMCID: PMC8506610 DOI: 10.1021/acsptsci.1c00094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 02/08/2023]
Abstract
Drug conjugates consisting of an antineoplastic drug and a targeting receptor ligand could be effective to overcome the heavy side effects of unselective anticancer agents. To address this need, we report here the results of a project aimed to study agonist and antagonist integrin ligands as targeting head of molecular cargoes for the selective delivery of 5-fluorouracil (5-FU) to cancer or noncancer cells. Initially, two fluorescent β-lactam-based integrin ligands were synthesized and tested for an effective and selective internalization mediated by α4β1 or α5β1 integrins in Jurkat and K562 cells, respectively. No cellular uptake was observed for both fluorescent compounds in HEK293 noncancerous control cells. Afterward, three conjugates composed of the β-lactam-based integrin ligand, suitable linkers, and 5-FU were realized. The best compound E, acting as α5β1 integrin agonist, is able to selectively deliver 5-FU into tumor cells, successfully leading to cancer cell death.
Collapse
Affiliation(s)
- Monica Baiula
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Martina Cirillo
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giulia Martelli
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | | | - Elisa Gasparini
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | | | - Santi Mario Spampinato
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Daria Giacomini
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| |
Collapse
|
45
|
Li Z, Chen Q, Wang J, Pan X, Lu W. Research Progress and Application of Bioorthogonal Reactions in Biomolecular Analysis and Disease Diagnosis. Top Curr Chem (Cham) 2021; 379:39. [PMID: 34590223 DOI: 10.1007/s41061-021-00352-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022]
Abstract
Bioorthogonal reactions are rapid, specific and high yield reactions that can be performed in in vivo microenvironments or simulated microenvironments. At present, the main biorthogonal reactions include Staudinger ligation, copper-catalyzed azide alkyne cycloaddition, strain-promoted [3 + 2] reaction, tetrazine ligation, metal-catalyzed coupling reaction and photo-induced biorthogonal reactions. To date, many reviews have reported that bioorthogonal reactions have been used widely as a powerful tool in the field of life sciences, such as in target recognition, drug discovery, drug activation, omics research, visualization of life processes or exogenous bacterial infection processes, signal transduction pathway research, chemical reaction dynamics analysis, disease diagnosis and treatment. In contrast, to date, few studies have investigated the application of bioorthogonal reactions in the analysis of biomacromolecules in vivo. Therefore, the application of bioorthogonal reactions in the analysis of proteins, nucleic acids, metabolites, enzyme activities and other endogenous molecules, and the determination of disease-related targets is reviewed. In addition, this review discusses the future development opportunities and challenges of biorthogonal reactions. This review presents an overview of recent advances for application in biomolecular analysis and disease diagnosis, with a focus on proteins, metabolites and RNA detection.
Collapse
Affiliation(s)
- Zilong Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qinhua Chen
- Department of Pharmacy, Shenzhen Baoan Authentic TCM Therapy Hospital, Shenzhen, 518101, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wen Lu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
46
|
Šimon P, Tichotová M, García Gallardo M, Procházková E, Baszczyňski O. Phosphate-Based Self-Immolative Linkers for Tuneable Double Cargo Release. Chemistry 2021; 27:12763-12775. [PMID: 34058033 DOI: 10.1002/chem.202101805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Indexed: 12/18/2022]
Abstract
Phosphorus-based self-immolative (SI) linkers offer a wide range of applications, such as smart materials and drug-delivery systems. Phosphorus SI linkers are ideal candidates for double-cargo delivery platforms because they have a higher valency than carbon. A series of substituted phosphate linkers was designed for releasing two phenolic cargos through SI followed by chemical hydrolysis. Suitable modifications of the lactate spacer increased the cargo release rate significantly, from 1 day to 2 hours or 5 minutes, as shown for linkers containing p-fluoro phenol. In turn, double cargo linkers bearing p-methyl phenol released their cargo more slowly (4 days, 4 hours, and 15 minutes) than their p-fluoro analogues. The α-hydroxyisobutyrate linker released both cargos in 25 minutes. Our study expands the current portfolio of SI constructs by providing a double cargo delivery option, which is crucial to develop universal SI platforms.
Collapse
Affiliation(s)
- Petr Šimon
- Faculty of Science, Charles University, Prague, 128 43, Czech Republic
| | - Markéta Tichotová
- Faculty of Science, Charles University, Prague, 128 43, Czech Republic.,Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, 166 10, Czech Republic
| | | | - Eliška Procházková
- Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, 166 10, Czech Republic
| | - Ondřej Baszczyňski
- Faculty of Science, Charles University, Prague, 128 43, Czech Republic.,Institute of Organic Chemistry and Biochemistry, The Czech Academy of Sciences, Prague, 166 10, Czech Republic
| |
Collapse
|
47
|
Park SE, El-Sayed NS, Shamloo K, Lohan S, Kumar S, Sajid MI, Tiwari RK. Targeted Delivery of Cabazitaxel Using Cyclic Cell-Penetrating Peptide and Biomarkers of Extracellular Matrix for Prostate and Breast Cancer Therapy. Bioconjug Chem 2021; 32:1898-1914. [PMID: 34309357 DOI: 10.1021/acs.bioconjchem.1c00319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Targeted drug delivery for cancer therapy is an emerging area of research. Cancer cells overexpress certain biomarkers that can be exploited for their targeted therapy. Cyclic cell-penetrating peptides (cCPP) are increasingly assessed for intracellular cargo delivery in cancer cells. In this study, we have conjugated cabazitaxel (CBT) to the cCPP via an ester bond to assist CBT release in the tumor's acidic environment. Integrin targeting (RGDC, TP1) and extra domain B of fibronectin (EDB-Fn) targeting (CTVRTSAD, TP2) peptides were linked to the peptide-drug conjugate (cCPP-CBT) via a disulfide bond to provide targeting ability to the conjugates until they reach the tumor site. Conjugate 11 (TP1-cCPP-CBT) and conjugate 16 (TP2-cCPP-CBT) showed approximately 3-4-fold less antiproliferative activity on integrin and EDB-FN overexpressing cancer cell lines as compared to the CBT analogue used for comparison (CBT-GA, 5). Conjugates (11 and 16) were less toxic (31-34-fold less antiproliferative activity) to the normal human embryonic kidney (HEK-293) cells as compared to CBT. The flow cytometry and quantitative confocal microscopy data further confirm the selective efficacy of conjugates (TP1-cCPP-FAM (10) and TP1-cCPP-FAM (15)) toward biomarker overexpressing cancer cells. Furthermore, the stability and release studies of conjugate 11 revealed its therapeutic potential under different conditions, such as human plasma, different pHs, and redox conditions. This conjugation strategy was proven to enhance chemotherapeutics agents' efficacy and targeting and can be applied to other chemotherapeutic agents.
Collapse
Affiliation(s)
- Shang Eun Park
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Naglaa Salem El-Sayed
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States.,Cellulose and Paper Department, National Research Center, Dokki 12622, Cairo, Egypt
| | - Kiumars Shamloo
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Sandeep Lohan
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Sumit Kumar
- Department of Chemistry, Deenbandhu Chhotu Ram University of Science and Technology, Murthal, Haryana 131039, India
| | - Muhammad Imran Sajid
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States.,Faculty of Pharmacy, University of Central Punjab, Lahore 54000, Pakistan
| | - Rakesh Kumar Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| |
Collapse
|
48
|
Mckertish CM, Kayser V. Advances and Limitations of Antibody Drug Conjugates for Cancer. Biomedicines 2021; 9:872. [PMID: 34440076 PMCID: PMC8389690 DOI: 10.3390/biomedicines9080872] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022] Open
Abstract
The popularity of antibody drug conjugates (ADCs) has increased in recent years, mainly due to their unrivalled efficacy and specificity over chemotherapy agents. The success of the ADC is partly based on the stability and successful cleavage of selective linkers for the delivery of the payload. The current research focuses on overcoming intrinsic shortcomings that impact the successful development of ADCs. This review summarizes marketed and recently approved ADCs, compares the features of various linker designs and payloads commonly used for ADC conjugation, and outlines cancer specific ADCs that are currently in late-stage clinical trials for the treatment of cancer. In addition, it addresses the issues surrounding drug resistance and strategies to overcome resistance, the impact of a narrow therapeutic index on treatment outcomes, the impact of drug-antibody ratio (DAR) and hydrophobicity on ADC clearance and protein aggregation.
Collapse
Affiliation(s)
| | - Veysel Kayser
- Sydney School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|
49
|
Baraniak D, Boryski J. Triazole-Modified Nucleic Acids for the Application in Bioorganic and Medicinal Chemistry. Biomedicines 2021; 9:628. [PMID: 34073038 PMCID: PMC8229351 DOI: 10.3390/biomedicines9060628] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023] Open
Abstract
This review covers studies which exploit triazole-modified nucleic acids in the range of chemistry and biology to medicine. The 1,2,3-triazole unit, which is obtained via click chemistry approach, shows valuable and unique properties. For example, it does not occur in nature, constitutes an additional pharmacophore with attractive properties being resistant to hydrolysis and other reactions at physiological pH, exhibits biological activity (i.e., antibacterial, antitumor, and antiviral), and can be considered as a rigid mimetic of amide linkage. Herein, it is presented a whole area of useful artificial compounds, from the clickable monomers and dimers to modified oligonucleotides, in the field of nucleic acids sciences. Such modifications of internucleotide linkages are designed to increase the hybridization binding affinity toward native DNA or RNA, to enhance resistance to nucleases, and to improve ability to penetrate cell membranes. The insertion of an artificial backbone is used for understanding effects of chemically modified oligonucleotides, and their potential usefulness in therapeutic applications. We describe the state-of-the-art knowledge on their implications for synthetic genes and other large modified DNA and RNA constructs including non-coding RNAs.
Collapse
Affiliation(s)
- Dagmara Baraniak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland;
| | | |
Collapse
|
50
|
Edupuganti VVSR, Tyndall JDA, Gamble AB. Self-immolative Linkers in Prodrugs and Antibody Drug Conjugates in Cancer Treatment. Recent Pat Anticancer Drug Discov 2021; 16:479-497. [PMID: 33966624 DOI: 10.2174/1574892816666210509001139] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/09/2021] [Accepted: 03/02/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND The design of anti-cancer therapies with high anti-tumour efficacy and reduced toxicity continues to be challenging. Anti-cancer prodrug and antibody-drug-conjugate (ADC) strategies that can specifically and efficiently deliver cytotoxic compounds to cancer cells have been used to overcome some of the challenges. Key to the success of many of these strategies is a self-immolative linker, which after activation can release the drug payload. Various types of triggerable self-immolative linkers are used in prodrugs and ADCs to improve their efficacy and safety. OBJECTIVE Numerous patents have reported the significance of self-immolative linkers in prodrugs and ADCs in cancer treatment. Based on the recent patent literature, we summarise methods for designing the site-specific activation of non-toxic prodrugs and ADCs in order to improve selectivity for killing cancer cells. METHODS In this review, an integrated view of the potential use of prodrugs and ADCs in cancer treatment are provided. This review presents recent patents and related publications over the past ten years to 2020. RESULTS The recent patent literature has been summarised for a wide variety of self-immolative PABC linkers, which are cleaved by factors including responding to the difference between the extracellular and intracellular environments (pH, ROS, glutathione), by over-expressed enzymes (cathepsin, plasmin, β-glucuronidase) or bioorthogonal activation. The mechanism for self-immolation involves the linker undergoing a 1,4- or 1,6-elimination (via electron cascade) or intramolecular cyclisation to release cytotoxic drug at the targeted site. CONCLUSION This review provides the commonly used strategies from recent patent literature in the development of prodrugs based on targeted cancer therapy and antibody-drug conjugates, which show promising results in therapeutic applications.
Collapse
Affiliation(s)
| | - Joel D A Tyndall
- School of Pharmacy, University of Otago, Dunedin, 9054. New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, Dunedin, 9054. New Zealand
| |
Collapse
|