51
|
Cai T, Shi L, Guo H, Li R, Cao W, Shen L, Zhu M, Tao Y. Detection and Characterization of In Vitro Payload-Containing Catabolites of Noncleavable Antibody-Drug Conjugates by High-Resolution Mass Spectrometry and Multiple Data Mining Tools. Drug Metab Dispos 2023; 51:591-598. [PMID: 36707253 DOI: 10.1124/dmd.122.001135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/28/2023] Open
Abstract
The formation and accumulation of payload-containing catabolites (PCCs) from a noncleavable antibody-drug conjugate (ADC) in targeted and normal tissues are directly associated with the therapeutic effect and toxicity of the ADC, respectively. Understanding the PCC formation is important for supporting the payload design and facilitating preclinical evaluation of ADCs. However, detection and identification of PCCs of a noncleavable ADC are challenging due to their low concentrations and unknown structures. The main objective of this study was to develop and apply a generic liquid chromatography-high-resolution mass spectrometry (LC-HRMS) method for profiling PCCs in vitro. Noncleavable ADCs, ado-trastuzumab emtansine (T-DM1) and ADC-1, were incubated in liver lysosomes, liver S9, and/or cancer cells followed by data acquisition using LC-HRMS. Profiling PCCs mainly relied on processing LC-HRMS datasets using untargeted precise and thorough background subtraction (PATBS) processing and targeted product ion filtering (PIF). As a result, 12 PCCs of T-DM1 were detected and structurally characterized in human liver lysosomal incubation, a majority of which consisted of 4-[N-maleimidomethyl]cyclohexane-1-carboxylate (MCC)-DM1 and a few amino acids. Additionally, the incubation of ADC-1 in human, rat, and monkey liver S9 and cancer cells generated one major and three very minor PCCs, verifying the payload design. The results demonstrate that PATBS enabled the comprehensive profiling of PCCs regardless of their molecular weights, charge states, and fragmentations. As a complementary tool, PIF detected specific PCCs with superior sensitivity. The combination of the in vitro metabolism systems and the LC-HRMS method is a useful approach to profiling in vitro PCCs of noncleavable ADCs in support of drug discovery programs. SIGNIFICANCE STATEMENT: Profiling in vitro payload-containing catabolites (PCCs) of a noncleavable antibody-drug conjugate (ADC) is important for optimization of the payload design and preclinical evaluation of ADC. However, currently used analytical approaches often fail to quickly provide reliable PCC profiling results. The work introduces a new liquid chromatography high resolution mass spectrometry method for comprehensive and rapid detection and characterization of PCCs released from a noncleavable ADC in liver lysosomes and S9 incubations.
Collapse
Affiliation(s)
- Tingting Cai
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Liqi Shi
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Huihui Guo
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Ruixing Li
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Weiqun Cao
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Liang Shen
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Mingshe Zhu
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Yi Tao
- Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Nanjing, Jiangsu, China (T.C.); Drug Metabolism and Pharmacokinetic Services, WuXi AppTec, Shanghai, China (L.S., R.L., W.C., L.S., Y.T.); Hangzhou DAC Biotechnology Co., Ltd., Hangzhou, China (H.G.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| |
Collapse
|
52
|
Lim JH, Park M, Park Y, Park SJ, Lee J, Hwang S, Lee J, Lee Y, Jo E, Shin YG. Evaluation of In Vivo Prepared Albumin-Drug Conjugate Using Immunoprecipitation Linked LC-MS Assay and Its Application to Mouse Pharmacokinetic Study. Molecules 2023; 28:3223. [PMID: 37049985 PMCID: PMC10096712 DOI: 10.3390/molecules28073223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
There have been many attempts in pharmaceutical industries and academia to improve the pharmacokinetic characteristics of anti-tumor small-molecule drugs by conjugating them with large molecules, such as monoclonal antibodies, called ADCs. In this context, albumin, one of the most abundant proteins in the blood, has also been proposed as a large molecule to be conjugated with anti-cancer small-molecule drugs. The half-life of albumin is 3 weeks in humans, and its distribution to tumors is higher than in normal tissues. However, few studies have been conducted for the in vivo prepared albumin-drug conjugates, possibly due to the lack of robust bioanalytical methods, which are critical for evaluating the ADME/PK properties of in vivo prepared albumin-drug conjugates. In this study, we developed a bioanalytical method of the albumin-conjugated MAC glucuronide phenol linked SN-38 ((2S,3S,4S,5R,6S)-6-(4-(((((((S)-4,11-diethyl-4-hydroxy-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano [3',4':6,7] indolizino [1,2-b] quinolin-9-yl)oxy)methyl)(2 (methylsulfonyl)ethyl)carbamoyl)oxy)methyl)-2-(2-(3-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1-yl)-N-methylpropanamido)acetamido)phenoxy)-3,4,5-trihydroxytetra-hydro-2H-pyran-2-carboxylic acid) as a proof-of-concept. This method is based on immunoprecipitation using magnetic beads and the quantification of albumin-conjugated drug concentration using LC-qTOF/MS in mouse plasma. Finally, the developed method was applied to the in vivo intravenous (IV) mouse pharmacokinetic study of MAC glucuronide phenol-linked SN-38.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Young G. Shin
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea; (J.-H.L.)
| |
Collapse
|
53
|
Kobzev D, Prasad C, Walunj D, Gotman H, Semenova O, Bazylevich A, Patsenker L, Gellerman G. Synthesis and biological evaluation of theranostic Trastuzumab–SN38 conjugate for Near-IR fluorescence imaging and targeted therapy of HER2+ breast cancer. Eur J Med Chem 2023; 252:115298. [PMID: 36966651 DOI: 10.1016/j.ejmech.2023.115298] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 04/03/2023]
Abstract
Here, we report on the design, synthesis, and biological evaluation of a new theranostic antibody drug conjugate (ADC), Cy5-Ab-SS-SN38, that consists of the HER2-specific antibody trastuzumab (Ab) connected to the near infrared (NIR) pentamethine cyanine dye Cy5 and SN38, which is a bioactive metabolite of the anticancer drug irinotecan. SN38 is bound to an antibody through a glutathione-responsive self-immolative disulfide carbamate linker. For the first time, we explored this linker in ADC and found that it to reduce the drug release rate, which is important for safe drug delivery. The developed ADC exhibited specific accumulation and nanomolar anti-breast cancer activity on HER2-positive (HER2+) cell lines but no effect on HER2-. Animals treated with this ADC exhibited good tolerance. In vivo studies have shown that the ADC had good targeting ability for HER2+ tumors with much higher anticancer potency than trastuzumab itself or a mixture of trastuzumab with SN38. Side-by-side HER2+/HER2-xenograft at the 10 mg/kg dose exhibited specific accumulation and reduction of HER2+ tumor but not accumulation or growth inhibition of HER2-counterpart. The self-immolative disulfide linker implemented in this study was proven to be successful, broadening its utilization with other antibodies for targeted anticancer therapy in general. We believe that the theranostic ADCs comprising the glutathione-responsive self-immolative disulfide carbamate linker are applicable for the treatment and fluorescent monitoring of malignancies and anticancer drug delivery.
Collapse
|
54
|
Postulating the possible cellular signalling mechanisms of antibody drug conjugates in Alzheimer's disease. Cell Signal 2023; 102:110539. [PMID: 36455831 DOI: 10.1016/j.cellsig.2022.110539] [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: 11/17/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders in the world. Although the basic pathology of the disease is elucidated, it is difficult to restore or prevent the worsening of neurodegeneration and its symptoms. Antibody and small molecule-based approaches have been studied and are in study individually, but a combined approach like conjugation has not been performed to date. The conjugation between antibodies and drugs which are already used for Alzheimer's treatment or developed specifically for this purpose may have better efficacy and dual action in mitigating Alzheimer's disease. A probable mechanism for antibody-drug conjugates in Alzheimer's disease is discussed in the present review.
Collapse
|
55
|
Huysamen A, Fadeyi OE, Mayuni G, Dogbey DM, Mungra N, Biteghe FAN, Hardcastle N, Ramamurthy D, Akinrinmade OA, Naran K, Cooper S, Lang D, Richter W, Hunter R, Barth S. Click Chemistry-Generated Auristatin F-Linker-Benzylguanine for a SNAP-Tag-Based Recombinant Antibody-Drug Conjugate Demonstrating Selective Cytotoxicity toward EGFR-Overexpressing Tumor Cells. ACS OMEGA 2023; 8:4026-4037. [PMID: 36743041 PMCID: PMC9893251 DOI: 10.1021/acsomega.2c06844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/03/2023] [Indexed: 06/18/2023]
Abstract
Antibody-drug conjugates (ADCs) are bifunctional molecules combining the targeting potential of monoclonal antibodies with the cancer-killing ability of cytotoxic drugs. This simple yet intelligently designed system directly addresses the lack of specificity encountered with conventional anti-cancer treatment regimes. However, despite their initial success, the generation of clinically sustainable and effective ADCs has been plagued by poor tumor penetration, undefined chemical linkages, unpredictable pharmacokinetic profiles, and heterogeneous mixtures of products. To this end, we generated a SNAP-tag-based fusion protein targeting the epidermal growth factor receptor (EGFR)-a biomarker of aggressive and drug-resistant cancers. Here, we demonstrate the use of a novel click coupling strategy to engineer a benzylguanine (BG)-linker-auristatin F (AuriF) piece that can be covalently tethered to the EGFR-targeting SNAP-tag-based fusion protein in an irreversible 1:1 stoichiometric reaction to form a homogeneous product. Furthermore, using these recombinant ADCs to target EGFR-overexpressing tumor cells, we provide a proof-of-principle for generating biologically active antimitotic therapeutic proteins capable of inducing cell death in a dose-dependent manner, thus alleviating some of the challenges of early ADC development.
Collapse
Affiliation(s)
- Allan
M. Huysamen
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Olaolu E. Fadeyi
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Grace Mayuni
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Dennis M. Dogbey
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Neelakshi Mungra
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- Centre
for Immunity and Immunotherapies, Seattle
Children’s Research Institute, Seattle, Washington 98101, United States
| | - Fleury A. N. Biteghe
- Department
of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, Los Angeles, California 90048, United States
| | - Natasha Hardcastle
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Dharanidharan Ramamurthy
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Olusiji A. Akinrinmade
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- Department
of Molecular Pharmacology, Albert Einstein
College of Medicine, Bronx, New York 10461, United States
| | - Krupa Naran
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Susan Cooper
- Division
of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | - Dirk Lang
- Division
of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | | | - Roger Hunter
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Stefan Barth
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- South
African Research Chair in Cancer Biotechnology, Department of Integrative
Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape
Town 7700, South Africa
| |
Collapse
|
56
|
Niegisch G. Antibody-Drug-Conjugates (ADC): A Novel Treatment Option in Urothelial Carcinoma. Methods Mol Biol 2023; 2684:293-301. [PMID: 37410242 DOI: 10.1007/978-1-0716-3291-8_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Antibody drug conjugates (ADC) are a new class of agents that have been expanding the spectrum of treatment options in metastatic urothelial carcinoma only recently. Preliminary data suggest that these compounds may have the potential even to replace current standard treatments as platinum-based chemotherapies. To this end, current and future preclinical and translational evaluation of novel treatment strategies should consider these novel compounds in addition to current standard options as well. In this context, the following article will provide an overview of this new class of agents, starting with general information on molecular structure and mode of action, clinical use of ADCs in urothelial carcinoma, and ending with considerations for designing preclinical and translational experiments implementing ADCs.
Collapse
Affiliation(s)
- Günter Niegisch
- Department of Urology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany.
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), CIO-D, Germany.
| |
Collapse
|
57
|
Liu L, Chen J. Therapeutic antibodies for precise cancer immunotherapy: current and future perspectives. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:555-569. [PMID: 37724258 PMCID: PMC10471122 DOI: 10.1515/mr-2022-0033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/25/2022] [Indexed: 09/20/2023]
Abstract
Antibodies, as one of the most important components of host adaptive immune system, play an important role in defense of infectious disease, immune surveillance, and autoimmune disease. Due to the development of recombinant antibody technology, antibody therapeutics become the largest and rapidly expanding drug to provide major health benefits to patients, especially for the treatment of cancer patients. Many antibody-based therapeutic strategies have been developed including monoclonal antibodies, antibody-drug conjugates, bispecific and trispecific antibodies and pro-antibodies with promising results from both clinical and pre-clinical trials. However, the response rate and side-effect still vary between patients with undefined mechanisms. Here, we summarized the current and future perspectives of antibody-based cancer immunotherapeutic strategies for designing next-generation drugs.
Collapse
Affiliation(s)
- Longchao Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiahui Chen
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
58
|
Antibody-Drug Conjugates in Myeloid Leukemias. Cancer J 2022; 28:454-461. [DOI: 10.1097/ppo.0000000000000635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
59
|
Marei HE, Cenciarelli C, Hasan A. Potential of antibody-drug conjugates (ADCs) for cancer therapy. Cancer Cell Int 2022; 22:255. [PMID: 35964048 PMCID: PMC9375290 DOI: 10.1186/s12935-022-02679-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
The primary purpose of ADCs is to increase the efficacy of anticancer medications by minimizing systemic drug distribution and targeting specific cells. Antibody conjugates (ADCs) have changed the way cancer is treated. However, because only a tiny fraction of patients experienced long-term advantages, current cancer preclinical and clinical research has been focused on combination trials. The complex interaction of ADCs with the tumor and its microenvironment appear to be reliant on the efficacy of a certain ADC, all of which have significant therapeutic consequences. Several clinical trials in various tumor types are now underway to examine the potential ADC therapy, based on encouraging preclinical results. This review tackles the potential use of ADCs in cancer therapy, emphasizing the essential processes underlying their positive therapeutic impacts on solid and hematological malignancies. Additionally, opportunities are explored to understand the mechanisms of ADCs action, the mechanism of resistance against ADCs, and how to overcome potential resistance following ADCs administration. Recent clinical findings have aroused interest, leading to a large increase in the number of ADCs in clinical trials. The rationale behind ADCs, as well as their primary features and recent research breakthroughs, will be discussed. We then offer an approach for maximizing the potential value that ADCs can bring to cancer patients by highlighting key ideas and distinct strategies.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
60
|
Cheng-Sánchez I, Moya-Utrera F, Porras-Alcalá C, López-Romero JM, Sarabia F. Antibody-Drug Conjugates Containing Payloads from Marine Origin. Mar Drugs 2022; 20:md20080494. [PMID: 36005497 PMCID: PMC9410405 DOI: 10.3390/md20080494] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are an important class of therapeutics for the treatment of cancer. Structurally, an ADC comprises an antibody, which serves as the delivery system, a payload drug that is a potent cytotoxin that kills cancer cells, and a chemical linker that connects the payload with the antibody. Unlike conventional chemotherapy methods, an ADC couples the selective targeting and pharmacokinetic characteristics related to the antibody with the potent cytotoxicity of the payload. This results in high specificity and potency by reducing off-target toxicities in patients by limiting the exposure of healthy tissues to the cytotoxic drug. As a consequence of these outstanding features, significant research efforts have been devoted to the design, synthesis, and development of ADCs, and several ADCs have been approved for clinical use. The ADC field not only relies upon biology and biochemistry (antibody) but also upon organic chemistry (linker and payload). In the latter, total synthesis of natural and designed cytotoxic compounds, together with the development of novel synthetic strategies, have been key aspects of the consecution of clinical ADCs. In the case of payloads from marine origin, impressive structural architectures and biological properties are observed, thus making them prime targets for chemical synthesis and the development of ADCs. In this review, we explore the molecular and biological diversity of ADCs, with particular emphasis on those containing marine cytotoxic drugs as the payload.
Collapse
Affiliation(s)
- Iván Cheng-Sánchez
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Correspondence:
| | - Federico Moya-Utrera
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| | - Cristina Porras-Alcalá
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| | - Juan M. López-Romero
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| | - Francisco Sarabia
- Department of Organic Chemistry, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain; (F.M.-U.); (C.P.-A.); (J.M.L.-R.); (F.S.)
| |
Collapse
|
61
|
Synthesis and Evaluation of Small Molecule Drug Conjugates Harnessing Thioester-Linked Maytansinoids. Pharmaceutics 2022; 14:pharmaceutics14071316. [PMID: 35890212 PMCID: PMC9323955 DOI: 10.3390/pharmaceutics14071316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 12/04/2022] Open
Abstract
Ligand-targeting drug conjugates are a class of clinically validated biopharmaceutical drugs constructed by conjugating cytotoxic drugs with specific disease antigen targeting ligands through appropriate linkers. The integrated linker-drug motif embedded within such a system can prevent the premature release during systemic circulation, thereby allowing the targeting ligand to engage with the disease antigen and selective accumulation. We have designed and synthesized new thioester-linked maytansinoid conjugates. By performing in vitro cytotoxicity, targeting ligand binding assay, and in vivo pharmacokinetic studies, we investigated the utility of this new linker-drug moiety in the small molecule drug conjugate (SMDC) system. In particular, we conjugated the thioester-linked maytansinoids to the phosphatidylserine-targeting small molecule zinc dipicolylamine and showed that Zn8_DM1 induced tumor regression in the HCC1806 triple-negative breast cancer xenograft model. Moreover, in a spontaneous sorafenib-resistant liver cancer model, Zn8_DM1 exhibited potent antitumor growth efficacy. From quantitative mRNA analysis of Zn8_DM1 treated-tumor tissues, we observed the elevation of gene expressions associated with a “hot inflamed tumor” state. With the identification and validation of a plethora of cancer-associated antigens in the “omics” era, this work provided the insight that antibody- or small molecule-based targeting ligands can be conjugated similarly to generate new ligand-targeting drug conjugates.
Collapse
|
62
|
Debnath U, Verma S, Patra J, Mandal SK. A review on recent synthetic routes and computational approaches for antibody drug conjugation developments used in anti-cancer therapy. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
63
|
Cavaco M, Castanho MARB, Neves V. The Use of Antibody-Antibiotic Conjugates to Fight Bacterial Infections. Front Microbiol 2022; 13:835677. [PMID: 35330773 PMCID: PMC8940529 DOI: 10.3389/fmicb.2022.835677] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/14/2022] [Indexed: 12/26/2022] Open
Abstract
The emergence of antimicrobial resistance (AMR) is rapidly increasing and it is one of the significant twenty-first century's healthcare challenges. Unfortunately, the development of effective antimicrobial agents is a much slower and complex process compared to the spread of AMR. Consequently, the current options in the treatment of AMR are limited. One of the main alternatives to conventional antibiotics is the use of antibody-antibiotic conjugates (AACs). These innovative bioengineered agents take advantage of the selectivity, favorable pharmacokinetic (PK), and safety of antibodies, allowing the administration of more potent antibiotics with less off-target effects. Although AACs' development is challenging due to the complexity of the three components, namely, the antibody, the antibiotic, and the linker, some successful examples are currently under clinical studies.
Collapse
Affiliation(s)
| | | | - Vera Neves
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
64
|
Sharma A, Balda S, Capalash N, Sharma P. Engineering multifunctional enzymes for agro-biomass utilization. BIORESOURCE TECHNOLOGY 2022; 347:126706. [PMID: 35033642 DOI: 10.1016/j.biortech.2022.126706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 06/14/2023]
Abstract
Lignocellulosic biomass is a plentiful renewable resource that can be converted into a wide range of high-value-added industrial products. However, the complexity of its structural integrity is one of the major constraints and requires combinations of different fibrolytic enzymes for the cost-effective, industrially and environmentally feasible transformation. An interesting approach is constructing multifunctional enzymes, either in a single polypeptide or by joining multiple domains with linkers and performing diverse reactions simultaneously, in a single host. The production of such chimera proteins multiplies the advantages of different enzymatic reactions in a single setup, in lesser time, at lower production cost and with desirable and improved catalytic activities. This review embodies the various domain-tailoring and extracellular secretion strategies, possible solutions to their challenges, and efforts to experimentally connect different catalytic activities in a single host, as well as their applications.
Collapse
Affiliation(s)
- Aarjoo Sharma
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Sanjeev Balda
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Neena Capalash
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Prince Sharma
- Department of Microbiology, Panjab University, Chandigarh, India.
| |
Collapse
|
65
|
Kramlinger VM, Dalvie D, Heck CJS, Kalgutkar AS, O'Neill J, Su D, Teitelbaum AM, Totah RA. Future of Biotransformation Science in the Pharmaceutical Industry. Drug Metab Dispos 2022; 50:258-267. [PMID: 34921097 DOI: 10.1124/dmd.121.000658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/23/2021] [Indexed: 11/22/2022] Open
Abstract
Over the past decades, the number of scientists trained in departments dedicated to traditional medicinal chemistry, biotransformation and/or chemical toxicology have seemingly declined. Yet, there remains a strong demand for such specialized skills in the pharmaceutical industry, particularly within drug metabolism/pharmacokinetics (DMPK) departments. In this position paper, the members of the Biotransformation, Mechanisms, and Pathways Focus Group (BMPFG) steering committee reflect on the diverse roles and responsibilities of scientists trained in the biotransformation field in pharmaceutical companies and contract research organizations. The BMPFG is affiliated with the International Society for the Study of Xenobiotics (ISSX) and was specifically created to promote the exchange of ideas pertaining to topics of current and future interest involving the metabolism of xenobiotics (including drugs). The authors also delve into the relevant education and diverse training skills required to successfully nurture the future cohort of industry biotransformation scientists and guide them toward a rewarding career path. The ability of scientists with a background in biotransformation and organic chemistry to creatively solve complex drug metabolism problems encountered during research and development efforts on both small and large molecular modalities is exemplified in five relevant case studies. Finally, the authors stress the importance and continued commitment to training the next generation of biotransformation scientists who are not only experienced in the metabolism of conventional small molecule therapeutics, but are also equipped to tackle emerging challenges associated with new drug discovery modalities including peptides, protein degraders, and antibodies. SIGNIFICANCE STATEMENT: Biotransformation and mechanistic drug metabolism scientists are critical to advancing chemical entities through discovery and development, yet the number of scientists academically trained for this role is on the decline. This position paper highlights the continuing demand for biotransformation scientists and the necessity of nurturing creative ways to train them and guarantee the future growth of this field.
Collapse
Affiliation(s)
- Valerie M Kramlinger
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Deepak Dalvie
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Carley J S Heck
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Amit S Kalgutkar
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - James O'Neill
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Dian Su
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Aaron M Teitelbaum
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| | - Rheem A Totah
- Translational Medicine, Novartis Institutes for Biomedical Research, Inc., Cambridge, Massachusetts (V.M.K.)
- Bristol Myers Squibb, San Diego, California (D.D.)
- Medicine Design, Pfizer Worldwide Research, Groton, Connecticut (C.J.S.H.); Medicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts (A.S.K.); Charles River Laboratories Edinburgh Ltd, Tranent, Scotland (J.O.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (D.S.)
- Drug Metabolism and Pharmacokinetics Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (A.M.T.); and Medicinal Chemistry, University of Washington, Seattle, Washington (R.A.T.)
| |
Collapse
|
66
|
Usama SM, Marker SC, Caldwell DR, Patel NL, Feng Y, Kalen JD, St Croix B, Schnermann MJ. Targeted Fluorogenic Cyanine Carbamates Enable In Vivo Analysis of Antibody-Drug Conjugate Linker Chemistry. J Am Chem Soc 2021; 143:21667-21675. [PMID: 34928588 DOI: 10.1021/jacs.1c10482] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibody-drug conjugates (ADCs) are a rapidly emerging therapeutic platform. The chemical linker between the antibody and the drug payload plays an essential role in the efficacy and tolerability of these agents. New methods that quantitatively assess the cleavage efficiency in complex tissue settings could provide valuable insights into the ADC design process. Here we report the development of a near-infrared (NIR) optical imaging approach that measures the site and extent of linker cleavage in mouse models. This approach is enabled by a superior variant of our recently devised cyanine carbamate (CyBam) platform. We identify a novel tertiary amine-containing norcyanine, the product of CyBam cleavage, that exhibits a dramatically increased cellular signal due to an improved cellular permeability and lysosomal accumulation. The resulting cyanine lysosome-targeting carbamates (CyLBams) are ∼50× brighter in cells, and we find this strategy is essential for high-contrast in vivo targeted imaging. Finally, we compare a panel of several common ADC linkers across two antibodies and tumor models. These studies indicate that cathepsin-cleavable linkers provide dramatically higher tumor activation relative to hindered or nonhindered disulfides, an observation that is only apparent with in vivo imaging. This strategy enables quantitative comparisons of cleavable linker chemistries in complex tissue settings with implications across the drug delivery landscape.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Sierra C Marker
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Donald R Caldwell
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Nimit L Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
67
|
Parakh S, Nicolazzo J, Scott AM, Gan HK. Antibody Drug Conjugates in Glioblastoma - Is There a Future for Them? Front Oncol 2021; 11:718590. [PMID: 34926242 PMCID: PMC8678283 DOI: 10.3389/fonc.2021.718590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and fatal malignancy that despite decades of trials has limited therapeutic options. Antibody drug conjugates (ADCs) are composed of a monoclonal antibody which specifically recognizes a cellular surface antigen linked to a cytotoxic payload. ADCs have demonstrated superior efficacy and/or reduced toxicity in a range of haematological and solid tumors resulting in nine ADCs receiving regulatory approval. ADCs have also been explored in patients with brain tumours but with limited success to date. While earlier generations ADCs in glioma patients have had limited success and high toxicity, newer and improved ADCs characterised by low immunogenicity and more effective payloads have shown promise in a range of tumour types. These newer ADCs have also been tested in glioma patients, however, with mixed results. Factors affecting the effectiveness of ADCs to target the CNS include the blood brain barrier which acts as a physical and biochemical barrier, the pro-cancerogenic and immunosuppressive tumor microenvironment and tumour characteristics like tumour volume and antigen expression. In this paper we review the data regarding the ongoing the development of ADCs in glioma patients as well as potential strategies to overcome these barriers to maximise their therapeutic potential.
Collapse
Affiliation(s)
- Sagun Parakh
- Department of Medical Oncology, Austin Hospital, Heidelberg, VIC, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
| | - Joseph Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Heidelberg, VIC, Australia
| | - Hui Kong Gan
- Department of Medical Oncology, Austin Hospital, Heidelberg, VIC, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, Australia
- Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
68
|
Giese M, Davis PD, Woodman RH, Hermanson G, Pokora A, Vermillion M. Linker Architectures as Steric Auxiliaries for Altering Enzyme-Mediated Payload Release from Bioconjugates. Bioconjug Chem 2021; 32:2257-2267. [PMID: 34587447 DOI: 10.1021/acs.bioconjchem.1c00429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protease-activated prodrugs leverage the increased activity of proteases in the tumor microenvironment and the tight regulation in healthy tissues to provide selective activation of cytotoxins in the tumor while minimizing toxicity to normal tissues. One of the largest classes of protease-activated prodrugs are composed of therapeutic agents conjugated to macromolecular carriers via peptide motifs that are substrates for cathepsin B, and antibody-drug conjugates are one of the most successful designs within this class. However, many of these peptide motifs are also cleaved by extracellular enzymes such as elastase and carboxylesterase 1C. Additionally, some peptide sequences have little selectivity for other lysosomal cathepsins, which have also been found to have extracellular activity in normal physiological processes. A lack of selectivity or oversensitivity to other extracellular enzymes can lead to off-target release of the cytotoxic payload and subsequent toxicities. In this report, we describe an approach for modulating cathepsin-mediated release of the cytotoxic payload through steric shielding provided by the synergistic effects of appropriately designed hydrophilic linkers and the conjugated carrier. We prepared a fluorogenic model payload with a Val-Cit cleavable trigger and attached the trigger-payload to a variety of PEG-based linker architectures with different numbers of PEG arms (y), different numbers of ethylene oxide units in each arm (n), and different distances between the cleavable trigger and PEG branch point (D'). These linker-payloads were then used to prepare DAR2 conjugates with the cleavable triggers at three different distances (D) from the antibody, and cathepsin-mediated payload release was monitored with in vitro assays. The results show that structural variables of the linker architectures can be manipulated to effectively shield enzymatically labile trigger-payloads from enzymes with readily accessible binding sites, and may offer an additional strategy for balancing off-target and tumor-targeted payload release.
Collapse
Affiliation(s)
- Matthew Giese
- Quanta BioDesign, 7470 Montgomery Drive, Plain City, Ohio 43064, United States
| | - Paul D Davis
- Quanta BioDesign, 7470 Montgomery Drive, Plain City, Ohio 43064, United States
| | - Robert H Woodman
- Quanta BioDesign, 7470 Montgomery Drive, Plain City, Ohio 43064, United States
| | - Greg Hermanson
- Quanta BioDesign, 7470 Montgomery Drive, Plain City, Ohio 43064, United States
| | - Alex Pokora
- Quanta BioDesign, 7470 Montgomery Drive, Plain City, Ohio 43064, United States
| | - Melissa Vermillion
- Quanta BioDesign, 7470 Montgomery Drive, Plain City, Ohio 43064, United States
| |
Collapse
|