1
|
Ripoll M, Cahuzac H, Dovgan I, Ursuegui S, Neuberg P, Erb S, Cianférani S, Kichler A, Remy JS, Wagner A. Supramolecular Bioconjugation Strategy for Antibody-Targeted Delivery of siRNA. Bioconjug Chem 2024. [PMID: 39321037 DOI: 10.1021/acs.bioconjchem.4c00304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
RNA interference is a widely used biological process by which double-stranded RNA induces sequence-specific gene silencing by targeting mRNA for degradation. However, the physicochemical properties of siRNAs make their delivery extremely challenging, thus limiting their bioavailability at the target site. In this context, we developed a versatile and selective siRNA delivery system of a trastuzumab-conjugated nanocarrier. These immunoconjugates consist of the assembly by electrostatic interactions of an oligonucleotide-modified antibody with a cationic micelle for the targeted delivery of siRNA in HER2-overexpressing cancer cells. Results show that, when associated with the corresponding siRNA at the appropriate N/P ratio, our supramolecular assembly was able to efficiently induce luciferase and PLK-1 gene silencing in a cell-selective manner in vitro.
Collapse
Affiliation(s)
- Manon Ripoll
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Héloïse Cahuzac
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Igor Dovgan
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Sylvain Ursuegui
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Patrick Neuberg
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Stephane Erb
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, University of Strasbourg, 25 rue Becquerel, Strasbourg 67087, France
- IPHC, CNRS, UMR7178, University of Strasbourg, Strasbourg 67087, France
| | - Sarah Cianférani
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, University of Strasbourg, 25 rue Becquerel, Strasbourg 67087, France
- IPHC, CNRS, UMR7178, University of Strasbourg, Strasbourg 67087, France
| | - Antoine Kichler
- Université de Strasbourg, Institut National de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Biomaterials and Bioengineering, UMR_S 1121 INSERM/EMR 7003 CNRS, Faculté de Pharmacie, Illkirch 67401, France
| | - Jean-Serge Remy
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| | - Alain Wagner
- Bio-Functional Chemistry (UMR 7199), LabEx Medalis, University of Strasbourg, 74 Route du Rhin, Illkirch-Graffenstaden 67400, France
| |
Collapse
|
2
|
Kathad U, Biyani N, Peru y Colón De Portugal RL, Zhou J, Kochat H, Bhatia K. Expanding the repertoire of Antibody Drug Conjugate (ADC) targets with improved tumor selectivity and range of potent payloads through in-silico analysis. PLoS One 2024; 19:e0308604. [PMID: 39186767 PMCID: PMC11346940 DOI: 10.1371/journal.pone.0308604] [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: 02/07/2024] [Accepted: 07/28/2024] [Indexed: 08/28/2024] Open
Abstract
Antibody-Drug Conjugates (ADCs) have emerged as a promising class of targeted cancer therapeutics. Further refinements are essential to unlock their full potential, which is currently limited by a lack of validated targets and payloads. Essential aspects of developing effective ADCs involve the identification of surface antigens, ideally distinguishing target tumor cells from healthy types, uniformly expressed, accompanied by a high potency payload capable of selective targeting. In this study, we integrated transcriptomics, proteomics, immunohistochemistry and cell surface membrane datasets from Human Protein Atlas, Xenabrowser and Gene Expression Omnibus utilizing Lantern Pharma's proprietary AI platform Response Algorithm for Drug positioning and Rescue (RADR®). We used this in combination with evidence based filtering to identify ADC targets with improved tumor selectivity. Our analysis identified a set of 82 targets and a total of 290 target indication combinations for effective tumor targeting. We evaluated the impact of tumor mutations on target expression levels by querying 416 genes in the TCGA mutation database against 22 tumor subtypes. Additionally, we assembled a catalog of compounds to identify potential payloads using the NCI-Developmental Therapeutics Program. Our payload mining strategy classified 729 compounds into three subclasses based on GI50 values spanning from pM to 10 nM range, in combination with sensitivity patterns across 9 different cancer indications. Our results identified a diverse range of both targets and payloads, that can serve to facilitate multiple choices for precise ADC targeting. We propose an initial approach to identify suitable target-indication-payload combinations, serving as a valuable starting point for development of future ADC candidates.
Collapse
Affiliation(s)
- Umesh Kathad
- Lantern Pharma Inc., Dallas, TX, United States of America
| | - Neha Biyani
- Lantern Pharma Inc., Dallas, TX, United States of America
| | | | - Jianli Zhou
- Lantern Pharma Inc., Dallas, TX, United States of America
| | - Harry Kochat
- The University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Kishor Bhatia
- Lantern Pharma Inc., Dallas, TX, United States of America
| |
Collapse
|
3
|
Anderson TS, McCormick AL, Smith SL, Lowe DB. Modeling antibody drug conjugate potential using a granzyme B antibody fusion protein. BMC Biol 2024; 22:66. [PMID: 38486229 PMCID: PMC10941411 DOI: 10.1186/s12915-024-01860-x] [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: 09/11/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Antibody drug conjugates (ADCs) constitute a promising class of targeted anti-tumor therapeutics that harness the selectivity of monoclonal antibodies with the potency of cytotoxic drugs. ADC development is best suited to initially screening antibody candidates for desired properties that potentiate target cell cytotoxicity. However, validating and producing an optimally designed ADC requires expertise and resources not readily available to certain laboratories. RESULTS In this study, we propose a novel approach to help streamline the identification of potential ADC candidates by utilizing a granzyme B (GrB)-based antibody fusion protein (AFP) for preliminary screening. GrB is a non-immunogenic serine protease expressed by immune effector cells such as CD8 + T cells that induces apoptotic activity and can be leveraged for targeted cell killing. CONCLUSIONS Our innovative model allows critical antibody parameters (including target cell binding, internalization, and cytotoxic potential) to be more reliably evaluated in vitro through the creation of an ADC surrogate. Successful incorporation of this AFP could also significantly expand and enhance ADC development pre-clinically, ultimately leading to the accelerated translation of ADC therapies for patients.
Collapse
Affiliation(s)
- Trevor S Anderson
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Amanda L McCormick
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Savanna L Smith
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Devin B Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA.
| |
Collapse
|
4
|
Rizvi SF, Zhang L, Zhang H, Fang Q. Peptide-Drug Conjugates: Design, Chemistry, and Drug Delivery System as a Novel Cancer Theranostic. ACS Pharmacol Transl Sci 2024; 7:309-334. [PMID: 38357281 PMCID: PMC10863443 DOI: 10.1021/acsptsci.3c00269] [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: 10/04/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 02/16/2024]
Abstract
The emergence of peptide-drug conjugates (PDCs) that utilize target-oriented peptide moieties as carriers of cytotoxic payloads, interconnected with various cleavable/noncleavable linkers, resulted in the key-foundation of the new era of targeted therapeutics. They are capable of retaining the integrity of conjugates in the blood circulatory system as well as releasing the drugs at the tumor microenvironment. Other valuable advantages are specificity and selectivity toward targeted-receptors, higher penetration ability, and drug-loading capacity, making them a suitable candidate to play their vital role as promising carrier agents. In this review, we summarized the types of cell-targeting (CTPs) and cell-penetrating peptides (CPPs) that have broad applications in the advancement of targeted drug-delivery systems (DDS). Moreover, the techniques to overcome the limitations of peptide-chemistry for their extensive implementation to construct the PDCs. Besides this, the diversified breakthrough of linker chemistry, and ample knowledge of various cytotoxic payloads used in PDCs in recent years, as well as the mechanism of action of PDCs was critically discussed. The principal aim is to provide scattered and diversified knowledge in one place and to help researchers understand the pinching knots in the science of PDC development, also their progression toward a bright future for PDCs as novel theranostics in clinical practice.
Collapse
Affiliation(s)
- Syed Faheem
Askari Rizvi
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, and
Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
- Institute
of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, 54000, Punjab Pakistan
| | - Linjie Zhang
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| | - Haixia Zhang
- State
Key Laboratory of Applied Organic Chemistry, College of Chemistry
and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| | - Quan Fang
- Key
Laboratory of Preclinical Study for New Drugs of Gansu Province, and
Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu P.R. China
| |
Collapse
|
5
|
Douez E, Allard-Vannier E, Amar IAM, Jolivet L, Boursin F, Maisonial-Besset A, Witkowski T, Chezal JM, Colas C, Letast S, Auvert E, Denevault-Sabourin C, Aubrey N, Joubert N. Branched pegylated linker-auristatin to control hydrophobicity for the production of homogeneous minibody-drug conjugate against HER2-positive breast cancer. J Control Release 2024; 366:567-584. [PMID: 38215985 DOI: 10.1016/j.jconrel.2024.01.012] [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: 07/21/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Trastuzumab emtansine (Kadcyla®) was the first antibody-drug conjugate (ADC) approved by the Food and Drug Administration in 2013 against a solid tumor, and the first ADC to treat human epidermal growth factor receptor 2 positive (HER2+) breast cancer. However, this second generation ADC is burden by several limitations included heterogeneity, limited activity against heterogeneous tumor (regarding antigen expression) and suboptimal tumor penetration. To address this, different development strategies are oriented towards homogeneous conjugation, new drugs, optimized linkers and/or smaller antibody formats. To reach better developed next generation ADCs, a key parameter to consider is the management of the hydrophobicity associated with the linker-drug, increasing with and limiting the drug-to-antibody ratio (DAR) of the ADC. Here, an innovative branched pegylated linker was developed, to control the hydrophobicity of the monomethyl auristatin E (MMAE) and its cathepsin B-sensitive trigger. This branched pegylated linker-MMAE was then used for the efficient generation of internalizing homogeneous ADC of DAR 8 and minibody-drug conjugate of DAR 4, targeting HER2. Both immunoconjugates were then evaluated in vitro and in vivo on breast cancer models. Interestingly, this study highlighted that the minibody-MMAE conjugate of DAR 4 was the best immunoconjugate regarding in vitro cellular internalization and cytotoxicity, gamma imaging, ex vivo biodistribution profile in mice and efficient reduction of tumor size in vivo. These results are very promising and encourage us to explore further fragment-drug conjugate development.
Collapse
Affiliation(s)
- Emmanuel Douez
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France; Pharmacy Department, Tours University Hospital, F-37200 Tours, France
| | - Emilie Allard-Vannier
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France.
| | | | - Louis Jolivet
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Fanny Boursin
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Aurélie Maisonial-Besset
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Tiffany Witkowski
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Jean-Michel Chezal
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Cyril Colas
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France; UMR 7311 ICOA, CNRS, University of Orléans, F-45067 Orléans, France
| | - Stéphanie Letast
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France
| | - Etienne Auvert
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France
| | | | - Nicolas Aubrey
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Nicolas Joubert
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France.
| |
Collapse
|
6
|
Ahad A, K. Saeed H, del Solar V, López-Hernández JE, Michel A, Mathew J, Lewis JS, Contel M. Shifting the Antibody-Drug Conjugate Paradigm: A Trastuzumab-Gold-Based Conjugate Demonstrates High Efficacy against Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer Mouse Model. ACS Pharmacol Transl Sci 2023; 6:1972-1986. [PMID: 38093840 PMCID: PMC10714425 DOI: 10.1021/acsptsci.3c00270] [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: 10/04/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 01/17/2024]
Abstract
Antibody-drug conjugates (ADCs) combine the selectivity of monoclonal antibodies (mAbs) with the efficacy of chemotherapeutics to target cancers without toxicity to normal tissue. Clinically, most chemotherapeutic ADCs are based on complex organic molecules, while the conjugation of metallodrugs to mAbs has been overlooked, despite the resurgent interest in metal-based drugs as cancer chemotherapeutics. In 2019, we described the first gold ADCs containing gold-triphenylphosphane fragments as a proof of concept. The ADCs (based on the antibody trastuzumab) were selective and highly active against HER2-positive breast cancer cells. In this study, we developed site-specific ADCs (Thio-1b and Thio-2b) using the cysteine-engineered trastuzumab derivative THIOMAB antibody technology with gold(I)-containing phosphanes and a maleimide-based linker amenable to bioconjugation (1b and 2b). In addition, we developed lysine-directed ADCs with gold payloads based on phosphanes and N-heterocyclic carbenes featuring an activated ester moiety (2c and 5c) with trastuzumab (Tras-2c and Tras-5c) and another anti-HER2 antibody, pertuzumab (Per-2c and Per-5c). Both sets of ADCs demonstrated significant anticancer potency in vitro assays. Based on these results, one ADC (Tras-2c), containing the [Au(PEt3)] fragment present in FDA-approved auranofin, was selected for an in vivo antitumor efficacy study. Immunocompromised mice xenografted with the HER2-positive human cancer cell line SKBR-3 exhibited almost complete tumor reduction and low toxicity with intravenous administration of Tras-2c. With this highly selective targeting system, we demonstrated that a subnanomolar cytotoxicity profile in cells is not required for an impressive antitumor effect in a mouse xenograft model.
Collapse
Affiliation(s)
- Afruja Ahad
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Hiwa K. Saeed
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Virginia del Solar
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
| | - Javier E. López-Hernández
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| | - Alexa Michel
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
| | - Joshua Mathew
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
| | - Jason S. Lewis
- Department
of Radiology, Memorial Sloan Kettering Cancer
Center, New York, New York 10065, United States
- Molecular
Pharmacology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
- Radiochemistry
and Molecular Imaging Probes Core, Memorial
Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Maria Contel
- Department
of Chemistry, The City University of New
York, Brooklyn, New York 11210, United States
- Brooklyn
College Cancer Center, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States
- Biology
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Chemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
- Biochemistry
PhD Programs, The Graduate Center, The City
University of New York, New York, New York 10016, United States
| |
Collapse
|
7
|
Prange CJ, Hu X, Tang L. Smart chemistry for traceless release of anticancer therapeutics. Biomaterials 2023; 303:122353. [PMID: 37925794 DOI: 10.1016/j.biomaterials.2023.122353] [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: 07/11/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
In the design of delivery strategies for anticancer therapeutics, the controlled release of intact cargo at the destined tumor and metastasis locations is of particular importance. To this end, stimuli-responsive chemical linkers have been extensively investigated owing to their ability to respond to tumor-specific physiological stimuli, such as lowered pH, altered redox conditions, increased radical oxygen species and pathological enzymatic activities. To prevent premature action and off-target effects, anticancer therapeutics are chemically modified to be transiently inactivated, a strategy known as prodrug development. Prodrugs are reactivated upon stimuli-dependent release at the sites of interest. As most drugs and therapeutic proteins have the optimal activity when released from carriers in their native and original forms, traceless release mechanisms are increasingly investigated. In this review, we summarize the chemical toolkit for developing innovative traceless prodrug strategies for stimuli-responsive drug delivery and discuss the applications of these chemical modifications in anticancer treatment including cancer immunotherapy.
Collapse
Affiliation(s)
- Céline Jasmin Prange
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland
| | - Xile Hu
- Institute of Chemical Sciences and Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland; Institute of Materials Science & Engineering, EPFL, Lausanne, CH-1015, Switzerland.
| |
Collapse
|
8
|
Riccardi F, Dal Bo M, Macor P, Toffoli G. A comprehensive overview on antibody-drug conjugates: from the conceptualization to cancer therapy. Front Pharmacol 2023; 14:1274088. [PMID: 37790810 PMCID: PMC10544916 DOI: 10.3389/fphar.2023.1274088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody-Drug Conjugates (ADCs) represent an innovative class of potent anti-cancer compounds that are widely used in the treatment of hematologic malignancies and solid tumors. Unlike conventional chemotherapeutic drug-based therapies, that are mainly associated with modest specificity and therapeutic benefit, the three key components that form an ADC (a monoclonal antibody bound to a cytotoxic drug via a chemical linker moiety) achieve remarkable improvement in terms of targeted killing of cancer cells and, while sparing healthy tissues, a reduction in systemic side effects caused by off-tumor toxicity. Based on their beneficial mechanism of action, 15 ADCs have been approved to date by the market approval by the Food and Drug Administration (FDA), the European Medicines Agency (EMA) and/or other international governmental agencies for use in clinical oncology, and hundreds are undergoing evaluation in the preclinical and clinical phases. Here, our aim is to provide a comprehensive overview of the key features revolving around ADC therapeutic strategy including their structural and targeting properties, mechanism of action, the role of the tumor microenvironment and review the approved ADCs in clinical oncology, providing discussion regarding their toxicity profile, clinical manifestations and use in novel combination therapies. Finally, we briefly review ADCs in other pathological contexts and provide key information regarding ADC manufacturing and analytical characterization.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| |
Collapse
|