1
|
Yaseen AA, Tumey LN. Advances and challenges in immunosuppressive antibody drug conjugates. Eur J Med Chem 2025; 291:117576. [PMID: 40186891 DOI: 10.1016/j.ejmech.2025.117576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Since the approval of Mylotarg™ in 2000 for acute myeloid leukemia, antibody-drug conjugates (ADCs) have significantly advanced precision medicine, particularly for oncology applications. ADCs combine an antibody, a linker, and a payload to result in a targeted therapeutic that minimizes toxicity resulting from systemic drug exposure. This review explores the innovative application of ADC technology towards immunosuppressive therapeutics, primarily focusing on antibody-mediated delivery of glucocorticoids (GCs). Despite their potent anti-inflammatory effects, the clinical use of GCs is limited by adverse systemic effects including osteoporosis, high blood sugar, adrenal insufficiency, weight gain, and glaucoma. Therefore, targeted delivery via ADCs presents a promising strategy to enhance therapeutic efficacy while reducing toxicity. Herein, we review the current status of immune-suppressing ADC technology, starting with early investigations of CD163-targeted dexamethasone and moving to the design of ADCs employing next-generation ultra-potent GCs. Additionally, we will discuss the current status of anti-inflammatory ADCs that employ non-glucocorticoid immune-suppressive medications. Throughout, we will highlight preclinical and clinical data that serves to derisk and drive investment in this new therapeutic class. In parallel, we will focus on ADC design principles that illustrate the importance of careful selection of payload, linker, and conjugation technology in this emerging field.
Collapse
Affiliation(s)
- Aiman A Yaseen
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, PO Box 6000, Binghamton, NY, 13902-6000, USA
| | - L Nathan Tumey
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, PO Box 6000, Binghamton, NY, 13902-6000, USA.
| |
Collapse
|
2
|
Dixit T, Vaidya A, Ravindran S. Therapeutic potential of antibody-drug conjugates possessing bifunctional anti-inflammatory action in the pathogenies of rheumatoid arthritis. Arthritis Res Ther 2024; 26:216. [PMID: 39695738 PMCID: PMC11656801 DOI: 10.1186/s13075-024-03452-0] [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: 08/10/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
In an age where there is a remarkable upsurge in developing precision medicines, antibody-drug conjugates (ADCs) have emerged as a progressive therapeutic strategy. ADCs typically consist of monoclonal antibodies (mAb) conjugated to the cytotoxic payloads by utilizing a linker, combining the benefits of definitive target specificity of mAbs and potent killing impact of payload to achieve precise and efficient elimination of target cells. In addition to their well-established role in oncology, ADCs are currently demonstrating encouraging potential in addressing the unmet requirements in the treatment of autoimmune conditions such as rheumatoid arthritis (RA). Prevalent long-term autoimmune disease RA costs billions of dollars annually but still, there is a lack of precision-targeted therapeutics with minimal side effects. This review provides an overview of the RA pathogenesis, pre-existing therapies, and their limitations, the introduction of ADCs in RA treatment, the mechanism of ADCs, and a summary of ADCs in preclinical and clinical trials. Based on the literature we also propose a strategy in ADC synthesis, which may increase the efficiency in targeting multifactorial diseases like RA. We propose to utilize DMARDs (Disease-modifying anti-rheumatic drugs), the first-line treatment for RA, as a payload for ADC synthesis. DMARDs are the only class of medication that limits the disease progression, but their efficacy is limited due to off-target toxicities. Hence, utilizing them as payload will help to deliver them directly at the targeted site, reducing their off-target toxicity, which in turn will increase their efficiency in targeting disease. Also, as mAbs are not sufficient to achieve remission, they are given in combinations with DMARDs. Hence, synthesizing ADCs may reduce the multiple and higher dosages given to patients, which in turn may enhance patient compliance.
Collapse
Affiliation(s)
- Tanu Dixit
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Anuradha Vaidya
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India
| | - Selvan Ravindran
- Symbiosis School of Biological Sciences (SSBS), Faculty of Medical & Health Sciences, Symbiosis International (Deemed University), Lavale, Pune, 412115, India.
| |
Collapse
|
3
|
Zhao C, Zhang R, Yang H, Gao Y, Zou Y, Zhang X. Antibody-drug conjugates for non-small cell lung cancer: Advantages and challenges in clinical translation. Biochem Pharmacol 2024; 226:116378. [PMID: 38908529 DOI: 10.1016/j.bcp.2024.116378] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/03/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Lung cancer is the leading cause of cancer death, with non-small cell lung cancer (NSCLC) accounting for approximately 85 % of all lung cancers and having a poor treatment and prognosis. Conventional clinical chemotherapy and immunotherapy are challenged by systemic toxicity and drug resistance, so researchers are increasingly focusing on antibody-drug conjugate (ADC), an innovative concept combining chemotherapy and targeted therapy, in which a drug selectively binds to antigens on the surface of a tumor cell via antibodies, which internalize the ADC, and then transfers the ADC to the lysosome via the endosomes to degrade the drug and kill the tumor cell. Despite the promising nature of ADCs, no ADC product for any indication including NSCLC has been approved for marketing by the FDA to date. In this review, we summarize the main advantages of ADCs and discuss in depth the design of the most desirable ADCs for NSCLC therapy. In addition to preclinical studies, we focus on the current state of clinical research on ADCs as interventions for the treatment of NSCLC by summarizing real-time clinical trial data from ClinicalTrials.gov, and reasonably speculate on the direction of the design of future generations of ADCs.
Collapse
Affiliation(s)
- Chenyu Zhao
- Department of China Medical University, The Queen's University of Belfast Joint College, School of Pharmacy, China Medical University, Shenyang 110122, China; School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Ruihan Zhang
- Department of China Medical University, The Queen's University of Belfast Joint College, School of Pharmacy, China Medical University, Shenyang 110122, China; School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, Shenyang 110122, China
| | - Yiwei Gao
- Department of China Medical University, The Queen's University of Belfast Joint College, School of Pharmacy, China Medical University, Shenyang 110122, China; School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Ying Zou
- Department of Rehabilitation Centre, Shengjing Hospital of China Medical University, Shenyang 110122, Liaoning, China.
| | - Xudong Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China.
| |
Collapse
|
4
|
Huang Z, Braunstein Z, Chen J, Wei Y, Rao X, Dong L, Zhong J. Precision Medicine in Rheumatic Diseases: Unlocking the Potential of Antibody-Drug Conjugates. Pharmacol Rev 2024; 76:579-598. [PMID: 38622001 DOI: 10.1124/pharmrev.123.001084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/25/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
In the era of precision medicine, antibody-drug conjugates (ADCs) have emerged as a cutting-edge therapeutic strategy. These innovative compounds combine the precision of monoclonal antibodies with the potent cell-killing or immune-modulating abilities of attached drug payloads. This unique strategy not only reduces off-target toxicity but also enhances the therapeutic effectiveness of drugs. Beyond their well established role in oncology, ADCs are now showing promising potential in addressing the unmet needs in the therapeutics of rheumatic diseases. Rheumatic diseases, a diverse group of chronic autoimmune diseases with varying etiologies, clinical presentations, and prognoses, often demand prolonged pharmacological interventions, creating a pressing need for novel, efficient, and low-risk treatment options. ADCs, with their ability to precisely target the immune components, have emerged as a novel therapeutic strategy in this context. This review will provide an overview of the core components and mechanisms behind ADCs, a summary of the latest clinical trials of ADCs for the treatment of rheumatic diseases, and a discussion of the challenges and future prospects faced by the development of next-generation ADCs. SIGNIFICANCE STATEMENT: There is a lack of efficient and low-risk targeted therapeutics for rheumatic diseases. Antibody-drug conjugates, a class of cutting-edge therapeutic drugs, have emerged as a promising targeted therapeutic strategy for rheumatic disease. Although there is limited literature summarizing the progress of antibody-drug conjugates in the field of rheumatic disease, updating the advancements in this area provides novel insights into the development of novel antirheumatic drugs.
Collapse
Affiliation(s)
- Zhiwen Huang
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Zachary Braunstein
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Jun Chen
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Yingying Wei
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Xiaoquan Rao
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Lingli Dong
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Jixin Zhong
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| |
Collapse
|
5
|
Marvin CC, Hobson AD, McPherson MJ, Hayes ME, Patel MV, Schmidt DL, Li T, Randolph JT, Bischoff AK, Fitzgibbons J, Wang L, Wang L, Hernandez A, Jia Y, Goess CA, Bryant SH, Mathieu SL, Xu J. Anti-TNF Thioester Glucocorticoid Antibody-Drug Conjugate Fully Inhibits Inflammation with Minimal Effect on Systemic Corticosterone Levels in a Mouse Arthritis Model. J Med Chem 2024; 67:9495-9515. [PMID: 38780432 DOI: 10.1021/acs.jmedchem.4c00598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
We describe the discovery of a thioester-containing glucocorticoid receptor modulator (GRM) payload and the corresponding antibody-drug conjugate (ADC). Payload 6 was designed for rapid hepatic inactivation to minimize systemic exposure of nonconjugated GRM. Mouse PK indicated that 6 is cleared 10-fold more rapidly than a first-generation GRM payload, resulting in 10-fold lower exposure and 3-fold decrease in Cmax. The anti-mTNF conjugate ADC5 fully inhibited inflammation in mouse contact hypersensitivity with minimal effects on corticosterone, a biomarker for systemic GRM effects, at doses up to and including 100 mg/kg. Concomitant inhibition of P1NP suggests potential delivery to cells involved in the remodeling of bone, which may be a consequence of TNF-targeting or bystander payload effects. Furthermore, ADC5 fully suppressed inflammation in collagen-induced arthritis mouse model after one 10 mg/kg dose for 21 days. The properties of the anti-hTNF conjugate were suitable for liquid formulation and may enable subcutaneous dosing.
Collapse
Affiliation(s)
- Christopher C Marvin
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Adrian D Hobson
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Michael J McPherson
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Martin E Hayes
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Meena V Patel
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Diana L Schmidt
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Tongmei Li
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - John T Randolph
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Agnieszka K Bischoff
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Julia Fitzgibbons
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Lu Wang
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Lu Wang
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Axel Hernandez
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Ying Jia
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Christian A Goess
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Shaughn H Bryant
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Suzanne L Mathieu
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Jianwen Xu
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| |
Collapse
|
6
|
Liu H, Ji M, Xiao P, Gou J, Yin T, He H, Tang X, Zhang Y. Glucocorticoids-based prodrug design: Current strategies and research progress. Asian J Pharm Sci 2024; 19:100922. [PMID: 38966286 PMCID: PMC11222810 DOI: 10.1016/j.ajps.2024.100922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/04/2024] [Accepted: 03/06/2024] [Indexed: 07/06/2024] Open
Abstract
Attributing to their broad pharmacological effects encompassing anti-inflammation, antitoxin, and immunosuppression, glucocorticoids (GCs) are extensively utilized in the clinic for the treatment of diverse diseases such as lupus erythematosus, nephritis, arthritis, ulcerative colitis, asthma, keratitis, macular edema, and leukemia. However, long-term use often causes undesirable side effects, including metabolic disorders-induced Cushing's syndrome (buffalo back, full moon face, hyperglycemia, etc.), osteoporosis, aggravated infection, psychosis, glaucoma, and cataract. These notorious side effects seriously compromise patients' quality of life, especially in patients with chronic diseases. Therefore, glucocorticoid-based advanced drug delivery systems for reducing adverse effects have received extensive attention. Among them, prodrugs have the advantages of low investment, low risk, and high success rate, making them a promising strategy. In this review, we propose the strategies for the design and summarize current research progress of glucocorticoid-based prodrugs in recent decades, including polymer-based prodrugs, dendrimer-based prodrugs, antibody-drug conjugates, peptide-drug conjugates, carbohydrate-based prodrugs, aliphatic acid-based prodrugs and so on. Besides, we also raise issues that need to be focused on during the development of glucocorticoid-based prodrugs. This review is expected to be helpful for the research and development of novel GCs and prodrugs.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Muse Ji
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Peifu Xiao
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jingxin Gou
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Yin
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haibing He
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xing Tang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Zhang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
7
|
Osgood AO, Singha Roy SJ, Koo D, Gu R, Chatterjee A. A Genetically Encoded Photocaged Cysteine for Facile Site-Specific Introduction of Conjugation-Ready Thiol Residues in Antibodies. Bioconjug Chem 2024; 35:457-464. [PMID: 38548654 PMCID: PMC11789925 DOI: 10.1021/acs.bioconjchem.3c00513] [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] [Indexed: 04/18/2024]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a powerful class of anticancer therapeutics that enable the selective delivery of toxic payloads into target cells. There is increasing appreciation for the importance of synthesizing such ADCs in a defined manner where the payload is attached at specific permissive sites on the antibody with a defined drug to antibody ratio. Additionally, the ability to systematically alter the site of attachment is important to fine-tune the therapeutic properties of the ADC. Engineered cysteine residues have been used to achieve such site-specific programmable attachment of drug molecules onto antibodies. However, engineered cysteine residues on antibodies often get "disulfide-capped" during secretion and require reductive regeneration prior to conjugation. This reductive step also reduces structurally important disulfide bonds in the antibody itself, which must be regenerated through oxidation. This multistep, cumbersome process reduces the efficiency of conjugation and presents logistical challenges. Additionally, certain engineered cysteine sites are resistant to reductive regeneration, limiting their utility and the overall scope of this conjugation strategy. In this work, we utilize a genetically encoded photocaged cysteine residue that can be site-specifically installed into the antibody. This photocaged amino acid can be efficiently decaged using light, revealing a free cysteine residue available for conjugation without disrupting the antibody structure. We show that this ncAA can be incorporated at several positions within full-length recombinant trastuzumab and decaged efficiently. We further used this method to generate a functional ADC site-specifically modified with monomethyl auristatin F (MMAF).
Collapse
Affiliation(s)
- Arianna O. Osgood
- Department of Chemistry, Boston College, 2609 Beacon Street, 201 Merkert Chemistry Center, Chestnut Hill, Massachusetts 02467, United States
| | - Soumya Jyoti Singha Roy
- Department of Chemistry, Boston College, 2609 Beacon Street, 201 Merkert Chemistry Center, Chestnut Hill, Massachusetts 02467, United States
| | - David Koo
- Department of Chemistry, Boston College, 2609 Beacon Street, 201 Merkert Chemistry Center, Chestnut Hill, Massachusetts 02467, United States
| | - Renpeng Gu
- Department of Chemistry, Boston College, 2609 Beacon Street, 201 Merkert Chemistry Center, Chestnut Hill, Massachusetts 02467, United States
| | - Abhishek Chatterjee
- Department of Chemistry, Boston College, 2609 Beacon Street, 201 Merkert Chemistry Center, Chestnut Hill, Massachusetts 02467, United States
| |
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
|
McPherson MJ, Hobson AD, Hernandez A, Marvin CC, Waegell W, Goess C, Oh JZ, Shi D, Hayes ME, Wang L, Wang L, Schmidt D, Wang Z, Pitney V, McCarthy K, Jia Y, Wang C, Kang BN, Bryant S, Mathieu S, Ruzek M, Parmentier J, D'Cunha RR, Pang Y, Phillips L, Brown NJ, Xu J, Graff C, Tian Y, Longenecker KL, Qiu W, Zhu H, Liu W, Zheng P, Bi Y, Stoffel R. An anti-TNF-glucocorticoid receptor modulator antibody-drug conjugate is efficacious against immune-mediated inflammatory diseases. Sci Transl Med 2024; 16:eadd8936. [PMID: 38507467 DOI: 10.1126/scitranslmed.add8936] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 02/27/2024] [Indexed: 03/22/2024]
Abstract
Glucocorticoids (GCs) are efficacious drugs used for treating many inflammatory diseases, but the dose and duration of administration are limited because of severe side effects. We therefore sought to identify an approach to selectively target GCs to inflamed tissue. Previous work identified that anti-tumor necrosis factor (TNF) antibodies that bind to transmembrane TNF undergo internalization; therefore, an anti-TNF antibody-drug conjugate (ADC) would be mechanistically similar, where lysosomal catabolism could release a GC receptor modulator (GRM) payload to dampen immune cell activity. Consequently, we have generated an anti-TNF-GRM ADC with the aim of inhibiting pro-inflammatory cytokine production from stimulated human immune cells. In an acute mouse model of contact hypersensitivity, a murine surrogate anti-TNF-GRM ADC inhibited inflammatory responses with minimal effect on systemic GC biomarkers. In addition, in a mouse model of collagen-induced arthritis, single-dose administration of the ADC, delivered at disease onset, was able to completely inhibit arthritis for greater than 30 days, whereas an anti-TNF monoclonal antibody only partially inhibited disease. ADC treatment at the peak of disease was also able to attenuate the arthritic phenotype. Clinical data for a human anti-TNF-GRM ADC (ABBV-3373) from a single ascending dose phase 1 study in healthy volunteers demonstrated antibody-like pharmacokinetic profiles and a lack of impact on serum cortisol concentrations at predicted therapeutic doses. These data suggest that an anti-TNF-GRM ADC may provide improved efficacy beyond anti-TNF alone in immune mediated diseases while minimizing systemic side effects associated with standard GC treatment.
Collapse
Affiliation(s)
| | - Adrian D Hobson
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Axel Hernandez
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | | | - Wendy Waegell
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Christian Goess
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Jason Z Oh
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Dan Shi
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Martin E Hayes
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Lu Wang
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Lu Wang
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Diana Schmidt
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | - Zhi Wang
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | - Victoria Pitney
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | | | - Ying Jia
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Ce Wang
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Bit Na Kang
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Shaughn Bryant
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Suzanne Mathieu
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Melanie Ruzek
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Julie Parmentier
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | | | - Yinuo Pang
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | - Lucy Phillips
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Nathan J Brown
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Jianwen Xu
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Candace Graff
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Yu Tian
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | | | - Wei Qiu
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | - Haizhong Zhu
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, USA
| | - Wei Liu
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Pingping Zheng
- AbbVie Bay Area, 1000 Gateway Boulevard, South San Francisco, CA 94080, USA
| | - Yingtao Bi
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| | - Robert Stoffel
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA
| |
Collapse
|
10
|
Zhang A, Seiss K, Laborde L, Palacio-Ramirez S, Guthy D, Lanter M, Lorber J, Vulpetti A, Arista L, Zoller T, Radimerski T, Thoma C, Hebach C, Tschantz WR, Karpov A, Hollingworth GJ, D'Alessio JA, Ferretti S, Burger MT. Design, Synthesis, and In Vitro and In Vivo Evaluation of Cereblon Binding Bruton's Tyrosine Kinase (BTK) Degrader CD79b Targeted Antibody-Drug Conjugates. Bioconjug Chem 2024; 35:140-146. [PMID: 38265691 DOI: 10.1021/acs.bioconjchem.3c00535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Antibody-drug conjugates (ADCs) are an established modality that allow for targeted delivery of a potent molecule, or payload, to a desired site of action. ADCs, wherein the payload is a targeted protein degrader, are an emerging area in the field. Herein we describe our efforts of delivering a Bruton's tyrosine kinase (BTK) bifunctional degrader 1 via a CD79b mAb (monoclonal antibody) where the degrader is linked at the ligase binding portion of the payload via a cleavable linker to the mAb. The resulting CD79b ADCs, 3 and 4, exhibit in vitro degradation and cytotoxicity comparable with that of 1, and ADC 3 can achieve more sustained in vivo degradation than intravenously administered 1 with markedly reduced systemic exposure of the payload.
Collapse
Affiliation(s)
- Alan Zhang
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139 United States
| | - Katherine Seiss
- Oncology Biotherapeutics, Novartis Biomedical Research, Cambridge, Massachusetts 02139 United States
| | - Laurent Laborde
- Oncology, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Sebastian Palacio-Ramirez
- Novartis Biologics Center, Novartis Biomedical Research, Cambridge, Massachusetts 02139 United States
| | - Daniel Guthy
- Oncology, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Mylene Lanter
- Oncology, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Julien Lorber
- Global Discovery Chemistry, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Anna Vulpetti
- Global Discovery Chemistry, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Luca Arista
- Global Discovery Chemistry, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Thomas Zoller
- Global Discovery Chemistry, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | - Claudio Thoma
- Oncology, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Christina Hebach
- Oncology, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - William R Tschantz
- Novartis Biologics Center, Novartis Biomedical Research, Cambridge, Massachusetts 02139 United States
| | - Alexei Karpov
- Global Discovery Chemistry, Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | - Joseph A D'Alessio
- Oncology Biotherapeutics, Novartis Biomedical Research, Cambridge, Massachusetts 02139 United States
| | | | - Matthew T Burger
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139 United States
| |
Collapse
|
11
|
Sydenham JD, Seki H, Krajcovicova S, Zeng L, Schober T, Deingruber T, Spring DR. Site-selective peptide functionalisation mediated via vinyl-triazine linchpins. Chem Commun (Camb) 2024; 60:706-709. [PMID: 38108130 DOI: 10.1039/d3cc05213c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Herein we introduce 3-vinyl-1,2,4-triazines derivatives as dual-reactive linkers that exhibit selectivity towards cysteine and specific strained alkynes, enabling conjugate addition and inverse electron-demand Diels-Alder (IEDDA) reactions. This approach facilitates site-selective bioconjugation of biologically relevant peptides, followed by rapid and highly selective reactions with bicyclononyne (BCN) reagents.
Collapse
Affiliation(s)
- Jack D Sydenham
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.
| | - Hikaru Seki
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.
| | - Sona Krajcovicova
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.
- Department of Organic Chemistry, Palacky University in Olomouc, Tr. 17. Listopadu 12, Olomouc, Czech Republic
| | - Linwei Zeng
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.
| | - Tim Schober
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.
| | - Tomas Deingruber
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.
| | - David R Spring
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK.
| |
Collapse
|
12
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
13
|
Howe JM, Fang S, Watts KA, Xu F, Benjamin SR, Tumey LN. ValCitGlyPro-dexamethasone antibody conjugates selectively suppress the activation of human monocytes. RSC Med Chem 2023; 14:2348-2357. [PMID: 37974960 PMCID: PMC10650436 DOI: 10.1039/d3md00336a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/18/2023] [Indexed: 11/19/2023] Open
Abstract
Glucocorticoids (GCs) are effective in treating autoimmune and inflammatory disorders but come with significant side effects, many of which are mediated by non-immunological cells. Therefore, there is rapidly growing interest in using antibody drug conjugate (ADC) technology to deliver GCs specifically to immune cells, thereby minimizing off-target side effects. Herein, we report the study of anti-CD11a, anti-CD38, and anti-TNFα ADCs to deliver dexamethasone to monocytes. We found that anti-CD11a and anti-CD38 were rapidly internalized by monocytes, while uptake of anti-TNFα depended on pre-activation with LPS. Using these antibodies were attached to a novel linker system, ValCitGlyPro-Dex (VCGP-Dex), that efficiently released dexamethasone upon lysosomal catabolism. This linker relies on lysosomal cathepsins to cleave after the ValCit sequence, thereby releasing a GlyPro-Dex species that undergoes rapid self-immolation to form dexamethasone. The resulting monocyte-targeting ADCs bearing this linker payload effectively suppressed LPS-induced NFκB activation and cytokine release in both a monocytic cell line (THP1) and in human PBMCs. Anti-TNFα_VCGP-Dex and anti-CD38_VCGP-Dex were particularly effective, suppressing ∼60-80% of LPS-induced IL-6 release from PBMCs at 3-10 μg mL-1 concentrations. In contrast, the corresponding isotype control ADC (anti-RSV) and the corresponding naked antibodies (anti-CD38 and anti-TNFα) resulted in only modest suppression (0-30%) of LPS-induced IL-6. Taken together, these results provide further evidence of the ability of glucocorticoid-ADCs to selectively suppress immune responses, and highlight the potential of two targets (CD38 and TNFα) for the development of novel immune-suppressing ADCs.
Collapse
Affiliation(s)
- Justin M Howe
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University PO Box 6000 Binghamton NY 13902-6000 USA
| | - Siteng Fang
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University PO Box 6000 Binghamton NY 13902-6000 USA
| | - Kelsey A Watts
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University PO Box 6000 Binghamton NY 13902-6000 USA
| | - Fanny Xu
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University PO Box 6000 Binghamton NY 13902-6000 USA
| | - Samantha R Benjamin
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University PO Box 6000 Binghamton NY 13902-6000 USA
| | - L Nathan Tumey
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University PO Box 6000 Binghamton NY 13902-6000 USA
| |
Collapse
|
14
|
Abstract
For many years, antibody drug conjugates (ADC) have teased with the promise of targeted payload delivery to diseased cells, embracing the targeting of the antibody to which a cytotoxic payload is conjugated. During the past decade this promise has started to be realised with the approval of more than a dozen ADCs for the treatment of various cancers. Of these ADCs, brentuximab vedotin really laid the foundations of a template for a successful ADC with lysosomal payload release from a cleavable dipeptide linker, measured DAR by conjugation to the Cys-Cys interchain bonds of the antibody and a cytotoxic payload. Using this ADC design model oncology has now expanded their repertoire of payloads to include non-cytotoxic compounds. These new payload classes have their origins in prior medicinal chemistry programmes aiming to design selective oral small molecule drugs. While this may not have been achieved, the resulting compounds provide excellent starting points for ADC programmes with some compounds amenable to immediate linker attachment while for others extensive SAR and structural information offer invaluable design insights. Many of these new oncology payload classes are of interest to other therapeutic areas facilitating rapid access to drug-linkers for exploration as non-oncology ADCs. Other therapeutic areas have also pursued unique payload classes with glucocorticoid receptor modulators (GRM) being the most clinically advanced in immunology. Here, ADC payloads come full circle, as oncology is now investigating GRM payloads for the treatment of cancer. This chapter aims to cover all these new ADC approaches while describing the medicinal chemistry origins of the new non-cytotoxic payloads.
Collapse
Affiliation(s)
- Adrian D Hobson
- Small Molecule Therapeutics & Platform Technologies, AbbVie Bioresearch Center, Worcester, MA, United States.
| |
Collapse
|
15
|
Marvin CC, Hobson AD, McPherson M, Dunstan TA, Vargo TR, Hayes ME, Fettis MM, Bischoff A, Wang L, Wang L, Hernandez A, Jia Y, Oh JZ, Tian Y. Self-Immolative Carbamate Linkers for CD19-Budesonide Antibody-Drug Conjugates. Bioconjug Chem 2023; 34:1835-1850. [PMID: 37788373 DOI: 10.1021/acs.bioconjchem.3c00354] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Antibody-drug conjugates consist of potent small-molecule payloads linked to a targeting antibody. Payloads must possess a viable functional group by which a linker for conjugation can be attached. Linker-attachment options remain limited for the connection to payloads via hydroxyl groups. A releasing group based on 2-aminopyridine was developed to enable stable attachment of para-aminobenzyl carbamate (PABC) linkers to the C21-hydroxyl group of budesonide, a glucocorticoid receptor agonist. Payload release involves a cascade of two self-immolative events that are initiated by the protease-mediated cleavage of the dipeptide-PABC bond. Budesonide release rates were determined for a series of payload-linker intermediates in buffered solution at pH 7.4 and 5.4, leading to the identification of 2-aminopyridine as the preferred releasing group. Addition of a poly(ethylene glycol) group improved linker hydrophilicity, thereby providing CD19-budesonide ADCs with suitable properties. ADC23 demonstrated targeted delivery of budesonide to CD19-expressing cells and inhibited B-cell activation in mice.
Collapse
Affiliation(s)
- Christopher C Marvin
- AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Adrian D Hobson
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Michael McPherson
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Theresa A Dunstan
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Thomas R Vargo
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Martin E Hayes
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Margaret M Fettis
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Agnieszka Bischoff
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Lu Wang
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Lu Wang
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Axel Hernandez
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Ying Jia
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Jason Z Oh
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Yu Tian
- AbbVie Bioresearch Center, 381 Plantation Street, Worcester, Massachusetts 01605, United States
| |
Collapse
|
16
|
Rakotoarinoro N, Dyck YFK, Krebs SK, Assi MK, Parr MK, Stech M. A disruptive clickable antibody design for the generation of antibody-drug conjugates. Antib Ther 2023; 6:298-310. [PMID: 38107665 PMCID: PMC10720948 DOI: 10.1093/abt/tbad023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/05/2023] [Accepted: 10/14/2023] [Indexed: 12/19/2023] Open
Abstract
Background Antibody-drug conjugates are cancer therapeutics that combine specificity and toxicity. A highly cytotoxic drug is covalently attached to an antibody that directs it to cancer cells. The conjugation of the drug-linker to the antibody is a key point in research and development as well as in industrial production. The consensus is to conjugate the drug to a surface-exposed part of the antibody to ensure maximum conjugation efficiency. However, the hydrophobic nature of the majority of drugs used in antibody-drug conjugates leads to an increased hydrophobicity of the generated antibody-drug conjugates, resulting in higher liver clearance and decreased stability. Methods In contrast, we describe a non-conventional approach in which the drug is conjugated in a buried part of the antibody. To achieve this, a ready-to-click antibody design was created in which an azido-based non-canonical amino acid is introduced within the Fab cavity during antibody synthesis using nonsense suppression technology. The Fab cavity was preferred over the Fc cavity to circumvent issues related to cleavage of the IgG1 lower hinge region in the tumor microenvironment. Results This antibody design significantly increased the hydrophilicity of the generated antibody-drug conjugates compared to the current best-in-class designs based on non-canonical amino acids, while conjugation efficiency and functionality were maintained. The robustness of this native shielding effect and the versatility of this approach were also investigated. Conclusions This pioneer design may become a starting point for the improvement of antibody-drug conjugates and an option to consider for protecting drugs and linkers from unspecific interactions.
Collapse
Affiliation(s)
- Nathanaël Rakotoarinoro
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Yan F K Dyck
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Simon K Krebs
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
- Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
| | - Miriam-Kousso Assi
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
- Department of Biotechnology, Hamburg University of Applied Sciences, 21033 Hamburg, Germany
| | - Maria K Parr
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Marlitt Stech
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
| |
Collapse
|
17
|
Pal LB, Bule P, Khan W, Chella N. An Overview of the Development and Preclinical Evaluation of Antibody-Drug Conjugates for Non-Oncological Applications. Pharmaceutics 2023; 15:1807. [PMID: 37513995 PMCID: PMC10385119 DOI: 10.3390/pharmaceutics15071807] [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: 02/23/2023] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Typically, antibody-drug conjugates (ADCs) are made up of a humanized antibody and a small-molecule medication connected by a chemical linker. ADCs' ability to deliver cytotoxic agents to the specific site with reduced side effects showed promising results in oncology. To date, fourteen ADCs have been approved by the US Food and Drug Administration, and approximately 297 ADCs are in pre-clinical/clinical stages in the oncology area. Inspired by these outcomes, a few scientists explored the potential of antibody-drug conjugates in non-oncological conditions such as arthritis, myasthenia gravis, immunological disorders, and kidney failure. However, there are limited data available on the non-oncological applications of antibody-drug conjugates. This current review focuses on the non-oncological applications of antibody-drug conjugates, their developmental studies, testing procedures, in vitro evaluations, and pre-clinical testing. Additionally, a summary of the restrictions, difficulties, and prospects for ADCs in non-oncological applications is provided.
Collapse
Affiliation(s)
- Lal Bahadur Pal
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, Assam, India
| | - Prajakta Bule
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, Assam, India
| | - Wahid Khan
- Natco Research Centre, Natco Pharma Ltd., Hyderabad 500018, Telangana, India
| | - Naveen Chella
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, Assam, India
| |
Collapse
|
18
|
Negi A, Kesari KK, Voisin-Chiret AS. Estrogen Receptor-α Targeting: PROTACs, SNIPERs, Peptide-PROTACs, Antibody Conjugated PROTACs and SNIPERs. Pharmaceutics 2022; 14:pharmaceutics14112523. [PMID: 36432713 PMCID: PMC9699327 DOI: 10.3390/pharmaceutics14112523] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Targeting selective estrogen subtype receptors through typical medicinal chemistry approaches is based on occupancy-driven pharmacology. In occupancy-driven pharmacology, molecules are developed in order to inhibit the protein of interest (POI), and their popularity is based on their virtue of faster kinetics. However, such approaches have intrinsic flaws, such as pico-to-nanomolar range binding affinity and continuous dosage after a time interval for sustained inhibition of POI. These shortcomings were addressed by event-driven pharmacology-based approaches, which degrade the POI rather than inhibit it. One such example is PROTACs (Proteolysis targeting chimeras), which has become one of the highly successful strategies of event-driven pharmacology (pharmacology that does the degradation of POI and diminishes its functions). The selective targeting of estrogen receptor subtypes is always challenging for chemical biologists and medicinal chemists. Specifically, estrogen receptor α (ER-α) is expressed in nearly 70% of breast cancer and commonly overexpressed in ovarian, prostate, colon, and endometrial cancer. Therefore, conventional hormonal therapies are most prescribed to patients with ER + cancers. However, on prolonged use, resistance commonly developed against these therapies, which led to selective estrogen receptor degrader (SERD) becoming the first-line drug for metastatic ER + breast cancer. The SERD success shows that removing cellular ER-α is a promising approach to overcoming endocrine resistance. Depending on the mechanism of degradation of ER-α, various types of strategies of developed.
Collapse
Affiliation(s)
- Arvind Negi
- Department of Bioproduct and Biosystems, Aalto University, 00076 Espoo, Finland
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| | - Kavindra Kumar Kesari
- Department of Bioproduct and Biosystems, Aalto University, 00076 Espoo, Finland
- Department of Applied Physics, School of Science, Aalto University, 02150 Espoo, Finland
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| | - Anne Sophie Voisin-Chiret
- CERMN (Centre d’Etudes et de Recherche sur le Médicament de Normandie), Normandie University UNICAEN, 14000 Caen, France
- Correspondence: or (A.N.); or (K.K.K.); (A.S.V.-C.)
| |
Collapse
|
19
|
Dragovich PS. Antibody-Drug Conjugates for Immunology. J Med Chem 2022; 65:4496-4499. [PMID: 35285623 DOI: 10.1021/acs.jmedchem.2c00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The application of antibody-drug conjugates (ADCs) to fields outside of oncology is increasing but is still relatively uncommon. A recent publication describes the conjugation of glucocorticoid receptor modulators to antibodies as a means of improving the separation between desired anti-inflammatory activity and unwanted systemic side effects.
Collapse
Affiliation(s)
- Peter S Dragovich
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
20
|
Lee SJ, Kim S, Jo DH, Cho CS, Kim SR, Kang D, Chae J, Yoo DK, Ha S, Chung J, Kim JH. Specific ablation of PDGFRβ-overexpressing pericytes with antibody-drug conjugate potently inhibits pathologic ocular neovascularization in mouse models. COMMUNICATIONS MEDICINE 2021; 1:58. [PMID: 35602228 PMCID: PMC9053257 DOI: 10.1038/s43856-021-00059-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 11/10/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Crosstalk between pericytes and endothelial cells is critical for ocular neovascularization. Endothelial cells secrete platelet-derived growth factor (PDGF)-BB and recruit PDGF receptor β (PDGFRβ)-overexpressing pericytes, which in turn cover and stabilize neovessels, independent of vascular endothelial growth factor (VEGF). Therapeutic agents inhibiting PDGF-BB/PDGFRβ signaling were tested in clinical trials but failed to provide additional benefits over anti-VEGF agents. We tested whether an antibody-drug conjugate (ADC) - an engineered monoclonal antibody linked to a cytotoxic agent - could selectively ablate pericytes and suppress retinal and choroidal neovascularization. METHODS Immunoblotting, flow cytometry, cell viability test, and confocal microscopy were conducted to assess the internalization and cytotoxic effect of ADC targeting mPDGFRβ in an in vitro setting. Immunofluorescence staining of whole-mount retinas and retinal pigment epithelium-choroid-scleral complexes, electroretinography, and OptoMotry test were used to evaluate the effect and safety of ADC targeting mPDGFRβ in the mouse models of pathologic ocular neovascularization. RESULTS ADC targeting mPDGFRβ is effectively internalized into mouse brain vascular pericytes and showed significant cytotoxicity compared with the control ADC. We also show that specific ablation of PDGFRβ-overexpressing pericytes using an ADC potently inhibits pathologic ocular neovascularization in mouse models of oxygen-induced retinopathy and laser-induced choroidal neovascularization, while not provoking generalized retinal toxicity. CONCLUSION Our results suggest that removing PDGFRβ-expressing pericytes by an ADC targeting PDGFRβ could be a potential therapeutic strategy for pathologic ocular neovascularization.
Collapse
Affiliation(s)
- Seok Jae Lee
- grid.412484.f0000 0001 0302 820XFight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soohyun Kim
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.168010.e0000000419368956Present Address: Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305 USA ,grid.168010.e0000000419368956Present Address: Stanford ChEM-H, Stanford University, Stanford, CA 94305 USA
| | - Dong Hyun Jo
- grid.31501.360000 0004 0470 5905Department of Anatomy & Cell Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chang Sik Cho
- grid.412484.f0000 0001 0302 820XFight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Su Ree Kim
- grid.255649.90000 0001 2171 7754Department of Life Science, Fluorescence Core Imaging Center, Ewha Womans University, Seoul, Republic of Korea
| | - Dongmin Kang
- grid.255649.90000 0001 2171 7754Department of Life Science, Fluorescence Core Imaging Center, Ewha Womans University, Seoul, Republic of Korea
| | - Jisu Chae
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Duck Kyun Yoo
- grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Suji Ha
- grid.31501.360000 0004 0470 5905Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea ,grid.31501.360000 0004 0470 5905Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Junho Chung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Transplantation Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Jeong Hun Kim
- Fight against Angiogenesis-Related Blindness (FARB) Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Advanced Biomedical Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.
| |
Collapse
|
21
|
Clarisse D, Deng L, de Bosscher K, Lother A. Approaches towards tissue-selective pharmacology of the mineralocorticoid receptor. Br J Pharmacol 2021; 179:3235-3249. [PMID: 34698367 DOI: 10.1111/bph.15719] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/23/2021] [Accepted: 10/10/2021] [Indexed: 11/29/2022] Open
Abstract
Mineralocorticoid receptor antagonists (MRAs) are highly effective therapies for cardiovascular and renal disease. However, the widespread clinical use of currently available MRAs in cardiorenal medicine is hampered by an increased risk of hyperkalemia. The mineralocorticoid receptor (MR) is a nuclear receptor responsible for fluid and electrolyte homeostasis in epithelial tissues, whereas pathophysiological MR activation in nonepithelial tissues leads to undesirable pro-inflammatory and pro-fibrotic effects. Therefore, new strategies that selectively target the deleterious effects of MR but spare its physiological function are needed. In this review, we discuss recent pharmacological developments starting from novel non-steroidal MRAs that are now entering clinical use, such as finerenone or esaxerenone, to concepts arising from the current knowledge of the MR signaling pathway, aiming at receptor-coregulator interaction, epigenetics, or downstream effectors of MR.
Collapse
Affiliation(s)
- Dorien Clarisse
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
| | - Lisa Deng
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Karolien de Bosscher
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Translational Nuclear Receptor Research, VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Cardiology and Angiology I, University Heart Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
22
|
Theocharopoulos C, Lialios PP, Samarkos M, Gogas H, Ziogas DC. Antibody-Drug Conjugates: Functional Principles and Applications in Oncology and Beyond. Vaccines (Basel) 2021; 9:1111. [PMID: 34696218 PMCID: PMC8538104 DOI: 10.3390/vaccines9101111] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/28/2022] Open
Abstract
In the era of precision medicine, antibody-based therapeutics are rapidly enriched with emerging advances and new proof-of-concept formats. In this context, antibody-drug conjugates (ADCs) have evolved to merge the high selectivity and specificity of monoclonal antibodies (mAbs) with the cytotoxic potency of attached payloads. So far, ten ADCs have been approved by FDA for oncological indications and many others are currently being tested in clinical and preclinical level. This paper summarizes the essential components of ADCs, from their functional principles and structure up to their limitations and resistance mechanisms, focusing on all latest bioengineering breakthroughs such as bispecific mAbs, dual-drug platforms as well as novel linkers and conjugation chemistries. In continuation of our recent review on anticancer implication of ADC's technology, further insights regarding their potential usage outside of the oncological spectrum are also presented. Better understanding of immunoconjugates could maximize their efficacy and optimize their safety, extending their use in everyday clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Dimitrios C. Ziogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 115 27 Athens, Greece; (C.T.); (P.-P.L.); (M.S.); (H.G.)
| |
Collapse
|
23
|
Han A, Olsen O, D'Souza C, Shan J, Zhao F, Yanolatos J, Hovhannisyan Z, Haxhinasto S, Delfino F, Olson W. Development of Novel Glucocorticoids for Use in Antibody-Drug Conjugates for the Treatment of Inflammatory Diseases. J Med Chem 2021; 64:11958-11971. [PMID: 34378927 DOI: 10.1021/acs.jmedchem.1c00541] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glucocorticoids (GCs) are widely used to treat a variety of autoimmune and inflammatory diseases; however, systemic delivery of GCs is associated with side effects that affect essentially every organ system, reflecting the nearly ubiquitous expression of the glucocorticoid receptor (GR). Targeted delivery of GCs to diseased tissues using antibody-glucocorticoid conjugates (GC-ADCs) offers a therapeutic alternative to overcome these adverse effects. Herein, we describe novel classes of GCs that exhibited greater potency than dexamethasone and budesonide, a 100-fold selectivity toward the GR over other nuclear receptors, and no in vitro safety liability in pharmacology assays (hERG, AMES) and that demonstrated a substantial reduction in tumor necrosis factor-α (TNF-α) release in mice challenged with lipopolysaccharide (LPS). The site-specific conjugated GC-ADCs via cathepsin-cleavable linkers were highly stable in plasma and specifically released GCs in antigen-positive cells, suggesting that these novel GCs can serve as ADC payloads to treat autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Amy Han
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Olav Olsen
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | | | - Jing Shan
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Feng Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Jean Yanolatos
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | | | - Sokol Haxhinasto
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - Frank Delfino
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| | - William Olson
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York 10591, United States
| |
Collapse
|
24
|
The Chemistry Behind ADCs. Pharmaceuticals (Basel) 2021; 14:ph14050442. [PMID: 34067144 PMCID: PMC8152005 DOI: 10.3390/ph14050442] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/29/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023] Open
Abstract
Combining the selective targeting of tumor cells through antigen-directed recognition and potent cell-killing by cytotoxic payloads, antibody-drug conjugates (ADCs) have emerged in recent years as an efficient therapeutic approach for the treatment of various cancers. Besides a number of approved drugs already on the market, there is a formidable follow-up of ADC candidates in clinical development. While selection of the appropriate antibody (A) and drug payload (D) is dictated by the pharmacology of the targeted disease, one has a broader choice of the conjugating linker (C). In the present paper, we review the chemistry of ADCs with a particular emphasis on the medicinal chemistry perspective, focusing on the chemical methods that enable the efficient assembly of the ADC from its three components and the controlled release of the drug payload.
Collapse
|
25
|
Dragovich PS, Pillow TH, Blake RA, Sadowsky JD, Adaligil E, Adhikari P, Chen J, Corr N, Dela Cruz-Chuh J, Del Rosario G, Fullerton A, Hartman SJ, Jiang F, Kaufman S, Kleinheinz T, Kozak KR, Liu L, Lu Y, Mulvihill MM, Murray JM, O'Donohue A, Rowntree RK, Sawyer WS, Staben LR, Wai J, Wang J, Wei B, Wei W, Xu Z, Yao H, Yu SF, Zhang D, Zhang H, Zhang S, Zhao Y, Zhou H, Zhu X. Antibody-Mediated Delivery of Chimeric BRD4 Degraders. Part 2: Improvement of In Vitro Antiproliferation Activity and In Vivo Antitumor Efficacy. J Med Chem 2021; 64:2576-2607. [PMID: 33596073 DOI: 10.1021/acs.jmedchem.0c01846] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heterobifunctional compounds that direct the ubiquitination of intracellular proteins in a targeted manner via co-opted ubiquitin ligases have enormous potential to transform the field of medicinal chemistry. These chimeric molecules, often termed proteolysis-targeting chimeras (PROTACs) in the chemical literature, enable the controlled degradation of specific proteins via their direction to the cellular proteasome. In this report, we describe the second phase of our research focused on exploring antibody-drug conjugates (ADCs), which incorporate BRD4-targeting chimeric degrader entities. We employ a new BRD4-binding fragment in the construction of the chimeric ADC payloads that is significantly more potent than the corresponding entity utilized in our initial studies. The resulting BRD4-degrader antibody conjugates exhibit potent and antigen-dependent BRD4 degradation and antiproliferation activities in cell-based experiments. Multiple ADCs bearing chimeric BRD4-degrader payloads also exhibit strong, antigen-dependent antitumor efficacy in mouse xenograft assessments that employ several different tumor models.
Collapse
Affiliation(s)
- Peter S Dragovich
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Thomas H Pillow
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Robert A Blake
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jack D Sadowsky
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Emel Adaligil
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Pragya Adhikari
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jinhua Chen
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Nicholas Corr
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | | | - Aaron Fullerton
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Steven J Hartman
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Fan Jiang
- Viva Biotech, Structural Biology, 334 Aidisheng Road, Zhangjiang High-Tech Park, Shanghai 201203, China
| | - Susan Kaufman
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Tracy Kleinheinz
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Katherine R Kozak
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Liling Liu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ying Lu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Melinda M Mulvihill
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jeremy M Murray
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Aimee O'Donohue
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rebecca K Rowntree
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - William S Sawyer
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Leanna R Staben
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John Wai
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Jian Wang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - BinQing Wei
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Wentao Wei
- Viva Biotech, Structural Biology, 334 Aidisheng Road, Zhangjiang High-Tech Park, Shanghai 201203, China
| | - Zijin Xu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Hui Yao
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Shang-Fan Yu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Donglu Zhang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hongyan Zhang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Shenhua Zhang
- Viva Biotech, Structural Biology, 334 Aidisheng Road, Zhangjiang High-Tech Park, Shanghai 201203, China
| | - Yongxin Zhao
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Hao Zhou
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Xiaoyu Zhu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| |
Collapse
|
26
|
Dragovich PS, Pillow TH, Blake RA, Sadowsky JD, Adaligil E, Adhikari P, Bhakta S, Blaquiere N, Chen J, Dela Cruz-Chuh J, Gascoigne KE, Hartman SJ, He M, Kaufman S, Kleinheinz T, Kozak KR, Liu L, Liu L, Liu Q, Lu Y, Meng F, Mulvihill MM, O'Donohue A, Rowntree RK, Staben LR, Staben ST, Wai J, Wang J, Wei B, Wilson C, Xin J, Xu Z, Yao H, Zhang D, Zhang H, Zhou H, Zhu X. Antibody-Mediated Delivery of Chimeric BRD4 Degraders. Part 1: Exploration of Antibody Linker, Payload Loading, and Payload Molecular Properties. J Med Chem 2021; 64:2534-2575. [PMID: 33596065 DOI: 10.1021/acs.jmedchem.0c01845] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The biological and medicinal impacts of proteolysis-targeting chimeras (PROTACs) and related chimeric molecules that effect intracellular degradation of target proteins via ubiquitin ligase-mediated ubiquitination continue to grow. However, these chimeric entities are relatively large compounds that often possess molecular characteristics, which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. We therefore explored the conjugation of such molecules to monoclonal antibodies using technologies originally developed for cytotoxic payloads so as to provide alternate delivery options for these novel agents. In this report, we describe the first phase of our systematic development of antibody-drug conjugates (ADCs) derived from bromodomain-containing protein 4 (BRD4)-targeting chimeric degrader entities. We demonstrate the antigen-dependent delivery of the degrader payloads to PC3-S1 prostate cancer cells along with related impacts on MYC transcription and intracellular BRD4 levels. These experiments culminate with the identification of one degrader conjugate, which exhibits antigen-dependent antiproliferation effects in LNCaP prostate cancer cells.
Collapse
Affiliation(s)
- Peter S Dragovich
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Thomas H Pillow
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Robert A Blake
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jack D Sadowsky
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Emel Adaligil
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Pragya Adhikari
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Sunil Bhakta
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Nicole Blaquiere
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jinhua Chen
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | | | - Karen E Gascoigne
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Steven J Hartman
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Mingtao He
- Pharmaron Beijing, Co. Ltd., 6 Tai He Road, BDA, Beijing 100176, China
| | - Susan Kaufman
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Tracy Kleinheinz
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Katherine R Kozak
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Liang Liu
- Pharmaron Beijing, Co. Ltd., 6 Tai He Road, BDA, Beijing 100176, China
| | - Liling Liu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Qi Liu
- Pharmaron Beijing, Co. Ltd., 6 Tai He Road, BDA, Beijing 100176, China
| | - Ying Lu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Fanwei Meng
- Pharmaron Beijing, Co. Ltd., 6 Tai He Road, BDA, Beijing 100176, China
| | - Melinda M Mulvihill
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Aimee O'Donohue
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rebecca K Rowntree
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Leanna R Staben
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Steven T Staben
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John Wai
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Jian Wang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - BinQing Wei
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Catherine Wilson
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jianfeng Xin
- Pharmaron Beijing, Co. Ltd., 6 Tai He Road, BDA, Beijing 100176, China
| | - Zijin Xu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Hui Yao
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Donglu Zhang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hongyan Zhang
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Hao Zhou
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Xiaoyu Zhu
- WuXi AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| |
Collapse
|
27
|
Walsh SJ, Bargh JD, Dannheim FM, Hanby AR, Seki H, Counsell AJ, Ou X, Fowler E, Ashman N, Takada Y, Isidro-Llobet A, Parker JS, Carroll JS, Spring DR. Site-selective modification strategies in antibody-drug conjugates. Chem Soc Rev 2021; 50:1305-1353. [PMID: 33290462 DOI: 10.1039/d0cs00310g] [Citation(s) in RCA: 254] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates (ADCs) harness the highly specific targeting capabilities of an antibody to deliver a cytotoxic payload to specific cell types. They have garnered widespread interest in drug discovery, particularly in oncology, as discrimination between healthy and malignant tissues or cells can be achieved. Nine ADCs have received approval from the US Food and Drug Administration and more than 80 others are currently undergoing clinical investigations for a range of solid tumours and haematological malignancies. Extensive research over the past decade has highlighted the critical nature of the linkage strategy adopted to attach the payload to the antibody. Whilst early generation ADCs were primarily synthesised as heterogeneous mixtures, these were found to have sub-optimal pharmacokinetics, stability, tolerability and/or efficacy. Efforts have now shifted towards generating homogeneous constructs with precise drug loading and predetermined, controlled sites of attachment. Homogeneous ADCs have repeatedly demonstrated superior overall pharmacological profiles compared to their heterogeneous counterparts. A wide range of methods have been developed in the pursuit of homogeneity, comprising chemical or enzymatic methods or a combination thereof to afford precise modification of specific amino acid or sugar residues. In this review, we discuss advances in chemical and enzymatic methods for site-specific antibody modification that result in the generation of homogeneous ADCs.
Collapse
Affiliation(s)
- Stephen J Walsh
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Joubert N, Beck A, Dumontet C, Denevault-Sabourin C. Antibody-Drug Conjugates: The Last Decade. Pharmaceuticals (Basel) 2020; 13:ph13090245. [PMID: 32937862 PMCID: PMC7558467 DOI: 10.3390/ph13090245] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 01/01/2023] Open
Abstract
An armed antibody (antibody–drug conjugate or ADC) is a vectorized chemotherapy, which results from the grafting of a cytotoxic agent onto a monoclonal antibody via a judiciously constructed spacer arm. ADCs have made considerable progress in 10 years. While in 2009 only gemtuzumab ozogamicin (Mylotarg®) was used clinically, in 2020, 9 Food and Drug Administration (FDA)-approved ADCs are available, and more than 80 others are in active clinical studies. This review will focus on FDA-approved and late-stage ADCs, their limitations including their toxicity and associated resistance mechanisms, as well as new emerging strategies to address these issues and attempt to widen their therapeutic window. Finally, we will discuss their combination with conventional chemotherapy or checkpoint inhibitors, and their design for applications beyond oncology, to make ADCs the magic bullet that Paul Ehrlich dreamed of.
Collapse
Affiliation(s)
- Nicolas Joubert
- GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France;
- Correspondence:
| | - Alain Beck
- Institut de Recherche Pierre Fabre, Centre d’Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien en Genevois, France;
| | - Charles Dumontet
- Cancer Research Center of Lyon (CRCL), INSERM, 1052/CNRS 5286/UCBL, 69000 Lyon, France;
- Hospices Civils de Lyon, 69000 Lyon, France
| | - Caroline Denevault-Sabourin
- GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France;
| |
Collapse
|
29
|
New insights into the cell- and tissue-specificity of glucocorticoid actions. Cell Mol Immunol 2020; 18:269-278. [PMID: 32868909 PMCID: PMC7456664 DOI: 10.1038/s41423-020-00526-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/11/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids (GCs) are endogenous hormones that are crucial for the homeostasis of the organism and adaptation to the external environment. Because of their anti-inflammatory effects, synthetic GCs are also extensively used in clinical practice. However, almost all cells in the body are sensitive to GC regulation. As a result, these mediators have pleiotropic effects, which may be undesirable or detrimental to human health. Here, we summarize the recent findings that contribute to deciphering the molecular mechanisms downstream of glucocorticoid receptor activation. We also discuss the complex role of GCs in infectious diseases such as sepsis and COVID-19, in which the balance between pathogen elimination and protection against excessive inflammation and immunopathology needs to be tightly regulated. An understanding of the cell type- and context-specific actions of GCs from the molecular to the organismal level would help to optimize their therapeutic use.
Collapse
|
30
|
Anami Y, Tsuchikama K. Next-generation Antibody-drug Conjugates (ADCs): Exploring New Frontiers with Chemical Approaches. J SYN ORG CHEM JPN 2020. [DOI: 10.5059/yukigoseikyokaishi.78.503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | - Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston
| |
Collapse
|
31
|
Gauzy-Lazo L, Sassoon I, Brun MP. Advances in Antibody–Drug Conjugate Design: Current Clinical Landscape and Future Innovations. SLAS DISCOVERY 2020; 25:843-868. [DOI: 10.1177/2472555220912955] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The targeted delivery of potent cytotoxic molecules into cancer cells is considered a promising anticancer strategy. The design of clinically effective antibody–drug conjugates (ADCs), in which biologically active drugs are coupled through chemical linkers to monoclonal antibodies, has presented challenges for pharmaceutical researchers. After 30 years of intensive research and development activities, only seven ADCs have been approved for clinical use; two have received fast-track designation and two breakthrough therapy designation from the Food and Drug Administration. There is continued interest in the field, as documented by the growing number of candidates in clinical development. This review aims to summarize the most recent innovations that have been applied to the design of ADCs undergoing early- and late-stage clinical trials. Discovery and rational optimization of new payloads, chemical linkers, and antibody formats have improved the therapeutic index of next-generation ADCs, ultimately resulting in improved clinical benefit for the patients.
Collapse
Affiliation(s)
| | - Ingrid Sassoon
- Immuno-Oncology Therapeutic Area, Sanofi, Vitry-sur-Seine, France
| | | |
Collapse
|
32
|
Abstract
The advent of biologic therapies, particularly antibody therapeutics, has revolutionized the pharmacological treatment of many rheumatic diseases. Antibody discovery began with the immunization of mice for the production of rodent immunoglobulins, but advances in protein and genetic engineering have now made it possible to generate fully human antibodies, which are better tolerated by patients. For most clinical applications in rheumatology, antibodies have been used as blocking agents capable of neutralizing the function of pro-inflammatory proteins, such as TNF. The latest strategies involve antibody products armed with effector moieties, such as anti-inflammatory drugs or cytokines, or antibody products that are specific for multiple targets for the selective inhibition of inflammation at sites of disease. Antibodies are some of the best-selling drugs in the world, and with further advances in antibody development, engineering of armed antibodies and bispecific products will have an important role in the treatment of rheumatic diseases.
Collapse
|
33
|
Bargh JD, Walsh SJ, Isidro-Llobet A, Omarjee S, Carroll JS, Spring DR. Sulfatase-cleavable linkers for antibody-drug conjugates. Chem Sci 2020; 11:2375-2380. [PMID: 34084399 PMCID: PMC8157321 DOI: 10.1039/c9sc06410a] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/27/2020] [Indexed: 12/18/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a class of targeted drug delivery agents combining the cell-selectivity of monoclonal antibodies (mAbs) and the cytotoxicity of small molecules. These two components are joined by a covalent linker, whose nature is critical to the efficacy and safety of the ADC. Enzyme-cleavable dipeptidic linkers have emerged as a particularly effective ADC linker type due to their ability to selectively release the payload in the lysosomes of target cells. However, these linkers have a number of drawbacks, including instability in rodent plasma and their inherently high hydrophobicity. Here we show that arylsulfate-containing ADC linkers are cleaved by lysosomal sulfatase enzymes to tracelessly release their payload, while circumventing the instability problems associated with dipeptide-linkers. When incorporated with trastuzumab and the highly potent monomethyl auristatin E (MMAE) payload, the arylsulfate-containing ADC 2 and ADC 3 were more cytotoxic than the non-cleavable ADC 4 against HER2-positive cells, while maintaining selectivity over HER2-negative cells. We propose that the stability, solubility and synthetic tractability of our arylsulfate linkers make them an attractive new motif for cleavable ADC linkers, with clear benefits over the widely used dipeptidic linkers.
Collapse
Affiliation(s)
- Jonathan D Bargh
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| | - Stephen J Walsh
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge CB2 1EW UK
- Cancer Research UK Cambridge Institute, University of Cambridge Robinson Way Cambridge CB2 0RE UK
| | | | - Soleilmane Omarjee
- Cancer Research UK Cambridge Institute, University of Cambridge Robinson Way Cambridge CB2 0RE UK
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge Robinson Way Cambridge CB2 0RE UK
| | - David R Spring
- Department of Chemistry, University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| |
Collapse
|
34
|
Antibody Conjugates-Recent Advances and Future Innovations. Antibodies (Basel) 2020; 9:antib9010002. [PMID: 31936270 PMCID: PMC7148502 DOI: 10.3390/antib9010002] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/18/2022] Open
Abstract
Monoclonal antibodies have evolved from research tools to powerful therapeutics in the past 30 years. Clinical success rates of antibodies have exceeded expectations, resulting in heavy investment in biologics discovery and development in addition to traditional small molecules across the industry. However, protein therapeutics cannot drug targets intracellularly and are limited to soluble and cell-surface antigens. Tremendous strides have been made in antibody discovery, protein engineering, formulation, and delivery devices. These advances continue to push the boundaries of biologics to enable antibody conjugates to take advantage of the target specificity and long half-life from an antibody, while delivering highly potent small molecule drugs. While the "magic bullet" concept produced the first wave of antibody conjugates, these entities were met with limited clinical success. This review summarizes the advances and challenges in the field to date with emphasis on antibody conjugation, linker-payload chemistry, novel payload classes, absorption, distribution, metabolism, and excretion (ADME), and product developability. We discuss lessons learned in the development of oncology antibody conjugates and look towards future innovations enabling other therapeutic indications.
Collapse
|
35
|
Abstract
The majority of ADCs in preclinical and clinical development are for oncology indications where cytotoxic payloads are targeted to antigen-expressing cancer cells. However, the modulation of pathogenic cellular activity via ADC-mediated delivery of bioactive small molecules is also an attractive concept for non-oncology indications leading to an expanded application of the technology. Here we summarize those ADCs that have been described so far for non-oncology applications and which cover a variety of payload mechanisms beyond cell killing, from early in vitro proof-of-concept experiments to clinical trials. As our understanding of ADC technology continues to grow, it is anticipated that the development of ADCs as therapeutics for disease areas outside of oncology will also increase.
Collapse
Affiliation(s)
| | - Adrian D Hobson
- Abbvie Global Biologics, AbbVie Bioresearch Center, Worcester, MA, USA
| |
Collapse
|
36
|
Abstract
The prototypical ADC mechanism involving antigen-mediated uptake and lysosomal release is both elegantly simple and scientifically compelling. However, recent clinical-stage failures have prompted a reevaluation of this delivery paradigm and have resulted in an array of new technologies that have the potential to improve the safety and efficacy of up and coming programs. These innovations can generally be categorized into seven areas that will be elaborated on in this chapter: (1) Exploiting new payload mechanisms; (2) Increasing the drug-antibody ratio (DAR); (3) Increasing the antibody penetration; (4) Overcoming ADC resistance mechanisms; (5) Increasing the efficiency of ADC uptake and processing; (6) Mitigating off-target payload exposure; and (7) Employment of noncytotoxic payloads. It is our belief that these seven areas capture the current "landscape" of innovations that are taking place in the design of next-generation ADCs. Together, these advancements are reshaping the ADC field and providing a path forward in the face of the recent clinical setbacks.
Collapse
Affiliation(s)
- L Nathan Tumey
- Department of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA.
- Pfizer Inc., Groton, CT, USA.
| |
Collapse
|
37
|
Dragovich PS, Adhikari P, Blake RA, Blaquiere N, Chen J, Cheng YX, den Besten W, Han J, Hartman SJ, He J, He M, Rei Ingalla E, Kamath AV, Kleinheinz T, Lai T, Leipold DD, Li CS, Liu Q, Lu J, Lu Y, Meng F, Meng L, Ng C, Peng K, Lewis Phillips G, Pillow TH, Rowntree RK, Sadowsky JD, Sampath D, Staben L, Staben ST, Wai J, Wan K, Wang X, Wei B, Wertz IE, Xin J, Xu K, Yao H, Zang R, Zhang D, Zhou H, Zhao Y. Antibody-mediated delivery of chimeric protein degraders which target estrogen receptor alpha (ERα). Bioorg Med Chem Lett 2019; 30:126907. [PMID: 31902710 DOI: 10.1016/j.bmcl.2019.126907] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/31/2022]
Abstract
Chimeric molecules which effect intracellular degradation of target proteins via E3 ligase-mediated ubiquitination (e.g., PROTACs) are currently of high interest in medicinal chemistry. However, these entities are relatively large compounds that often possess molecular characteristics which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. Accordingly, we explored whether conjugation of chimeric degraders to monoclonal antibodies using technologies originally developed for cytotoxic payloads might provide alternate delivery options for these novel agents. In this report we describe the construction of several degrader-antibody conjugates comprised of two distinct ERα-targeting degrader entities and three independent ADC linker modalities. We subsequently demonstrate the antigen-dependent delivery to MCF7-neo/HER2 cells of the degrader payloads that are incorporated into these conjugates. We also provide evidence for efficient intracellular degrader release from one of the employed linkers. In addition, preliminary data are described which suggest that reasonably favorable in vivo stability properties are associated with the linkers utilized to construct the degrader conjugates.
Collapse
Affiliation(s)
| | - Pragya Adhikari
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Robert A Blake
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Jinhua Chen
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Yun-Xing Cheng
- Pharmaron Beijing, Co. Ltd., BDA Beijing, 6 Tai He Road, 100176, China
| | | | - Jinping Han
- Pharmaron Beijing, Co. Ltd., BDA Beijing, 6 Tai He Road, 100176, China
| | | | - Jintang He
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Mingtao He
- Pharmaron Beijing, Co. Ltd., BDA Beijing, 6 Tai He Road, 100176, China
| | | | - Amrita V Kamath
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Tommy Lai
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | | | - Chun Sing Li
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Qi Liu
- Pharmaron Beijing, Co. Ltd., BDA Beijing, 6 Tai He Road, 100176, China
| | - Jiawei Lu
- WuXi Biologics, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Ying Lu
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Fanwei Meng
- Pharmaron Beijing, Co. Ltd., BDA Beijing, 6 Tai He Road, 100176, China
| | - Lingyao Meng
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Carl Ng
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kaishan Peng
- WuXi Biologics, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | | | - Thomas H Pillow
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Jack D Sadowsky
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Deepak Sampath
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Leanna Staben
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Steven T Staben
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - John Wai
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Kunpeng Wan
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Xinxin Wang
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - BinQing Wei
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ingrid E Wertz
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jianfeng Xin
- Pharmaron Beijing, Co. Ltd., BDA Beijing, 6 Tai He Road, 100176, China
| | - Keyang Xu
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hui Yao
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Richard Zang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Donglu Zhang
- Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Hao Zhou
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| | - Yongxin Zhao
- WuXi AppTec, Waigaoqiao Free Trade Zone, 288 Fute Zhong Road, Shanghai 200131, China
| |
Collapse
|
38
|
Dal Corso A, Pignataro L, Belvisi L, Gennari C. Innovative Linker Strategies for Tumor‐Targeted Drug Conjugates. Chemistry 2019; 25:14740-14757. [DOI: 10.1002/chem.201903127] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/15/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Alberto Dal Corso
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Luca Pignataro
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Laura Belvisi
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Cesare Gennari
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| |
Collapse
|
39
|
Benjamin SR, Jackson CP, Fang S, Carlson DP, Guo Z, Tumey LN. Thiolation of Q295: Site-Specific Conjugation of Hydrophobic Payloads without the Need for Genetic Engineering. Mol Pharm 2019; 16:2795-2807. [PMID: 31067063 DOI: 10.1021/acs.molpharmaceut.9b00323] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Site-specific conjugation technology frequently relies on antibody engineering to incorporate rare or non-natural amino acids into the primary sequence of the protein. However, when the primary sequence is unknown or when antibody engineering is not feasible, there are very limited options for site-specific protein modification. We have developed a transglutaminase-mediated conjugation that incorporates a thiol at a "privileged" location on deglycosylated antibodies (Q295). Perhaps surprisingly, this conjugation employs a reported transglutaminase inhibitor, cystamine, as the key enzyme substrate. The chemical incorporation of a thiol at the Q295 site allows for the site-specific attachment of a plethora of commonly used and commercially available payloads via maleimide chemistry. Herein, we demonstrate the utility of this method by comparing the conjugatability, plasma stability, and in vitro potency of these site-specific antibody-drug conjugates (ADCs) with analogous endogenous cysteine conjugates. Cytotoxic ADCs prepared using this methodology are shown to exhibit comparable in vitro efficacy to stochastic cysteine conjugates while displaying dramatically improved plasma stability and conjugatability. In particular, we note that this technique appears to be useful for the incorporation of highly hydrophobic linker payloads without the addition of PEG modifiers. We postulate a possible mechanism for this feature by probing the local environment of the Q295 site with two fluorescent probes that are known to be sensitive to the local hydrophobic environment. In summary, we describe a highly practical method for the site-specific conjugation of genetically nonengineered antibodies, which results in plasma-stable ADCs with low intrinsic hydrophobicity. We believe that this technology will find broad utility in the ADC community.
Collapse
Affiliation(s)
- Samantha R Benjamin
- School of Pharmacy and Pharmaceutical Sciences , Binghamton University , P.O. Box 6000, Binghamton , New York 13902 , United States
| | - Courtney P Jackson
- School of Pharmacy and Pharmaceutical Sciences , Binghamton University , P.O. Box 6000, Binghamton , New York 13902 , United States
| | - Siteng Fang
- School of Pharmacy and Pharmaceutical Sciences , Binghamton University , P.O. Box 6000, Binghamton , New York 13902 , United States
| | - Dane P Carlson
- School of Pharmacy and Pharmaceutical Sciences , Binghamton University , P.O. Box 6000, Binghamton , New York 13902 , United States
| | - Zhongyuan Guo
- School of Pharmacy and Pharmaceutical Sciences , Binghamton University , P.O. Box 6000, Binghamton , New York 13902 , United States
| | - L Nathan Tumey
- School of Pharmacy and Pharmaceutical Sciences , Binghamton University , P.O. Box 6000, Binghamton , New York 13902 , United States
| |
Collapse
|
40
|
Luo SH, Yang K, Lin JY, Gao JJ, Wu XY, Wang ZY. Synthesis of amino acid derivatives of 5-alkoxy-3,4-dihalo-2(5H)-furanones and their preliminary bioactivity investigation as linkers. Org Biomol Chem 2019; 17:5138-5147. [PMID: 31073571 DOI: 10.1039/c9ob00736a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A series of amino acid derivatives are successfully synthesized via a metal-free C-N coupling reaction of 5-alkoxy-3,4-dihalo-2(5H)-furanones and amino acids. Their structures are well characterized with 1H NMR, 13C NMR, ESI-MS and elemental analysis. As potential linkers of the 2(5H)-furanone unit with other drug moieties containing a hydroxyl or amino group, the effect of amino acids is investigated by comparison with other 2(5H)-furanone compounds by constructing C-O/C-S bonds. The preliminary results of the biological activity assay by the MTT method on a series of cancer cell lines in vitro reveal that the introduction of amino acids basically has no toxic effect. This can lead to these 2(5H)-furanone derivatives being further well-linked with other bioactive moieties with amino or hydroxy groups as expected. Thus, the biological activity assay gives a direction for the design of bioactive 2(5H)-furanones based on these amino acid linkers.
Collapse
Affiliation(s)
- Shi-He Luo
- School of Chemistry and Environment, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, Guangzhou 510006, P. R. China. and School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou 510641, P. R. China
| | - Kai Yang
- School of Chemistry and Environment, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, Guangzhou 510006, P. R. China. and College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi province 341000, P. R. China.
| | - Jian-Yun Lin
- School of Chemistry and Environment, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, Guangzhou 510006, P. R. China.
| | - Juan-Juan Gao
- College of Sports and Rehabilitation, Gannan Medical University, Ganzhou, Jiangxi province 341000, P. R. China
| | - Xin-Yan Wu
- School of Chemistry and Environment, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, Guangzhou 510006, P. R. China.
| | - Zhao-Yang Wang
- School of Chemistry and Environment, South China Normal University, Key Laboratory of Theoretical Chemistry of Environment, Ministry of Education, Guangzhou Key Laboratory of Analytical Chemistry for Biomedicine, Guangzhou 510006, P. R. China. and School of Chemistry and Chemical Engineering, Key Laboratory of Functional Molecular Engineering of Guangdong Province, South China University of Technology, Guangzhou 510641, P. R. China
| |
Collapse
|