1
|
Schiavon E, Rezzola S, Filippi E, Turati M, Parrasia S, Bernardotto S, Stocco M, Szabò I, Mattarei A, Ronca R, Morpurgo M. A novel mertansine conjugate for acid-reversible targeted drug delivery validated through the Avidin-Nucleic-Acid-NanoASsembly platform. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102784. [PMID: 39236938 DOI: 10.1016/j.nano.2024.102784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/27/2024] [Accepted: 08/25/2024] [Indexed: 09/07/2024]
Abstract
In targeted cancer therapy, antibody-drug-conjugates using mertansine (DM1)-based cytotoxic compounds rely on covalent bonds for drug conjugation. Consequently, the cytotoxic DM1 derivative released upon their proteolytic digestion is up to 1000-fold less potent than DM1 and lacks a bystander effect. To overcome these limitations, we developed a DM1 derivative (keto-DM1) suitable for bioconjugation through an acid-reversible hydrazone bond. Its acid-reversible hydrazone conjugate with biotin (B-Hz-DM1) was generated and tested for efficacy using the cetuximab-targeted Avidin-Nucleic-Acid-NanoASsembly (ANANAS) nanoparticle (NP) platform. NP-tethered B-Hz-DM1 is stable at neutral pH and releases its active moiety only in endosome/lysosome mimicking acidic pH. In vitro, the NP/Cetux/B-Hz-DM1 assembly showed high potency on MDA-MB231 breast cancer cells. In vivo both B-Hz-DM1 and NP/Cetux/B-Hz-DM1 reduced tumor growth. A significantly major effect was exerted by the nanoformulation, associated with an increased in situ tumor cell death. Keto-DM1 is a promising acid-reversible mertansine derivative for targeted delivery in cancer therapy.
Collapse
Affiliation(s)
- Elisa Schiavon
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Sara Rezzola
- Molecular and Translational Medicine Dept (DMMT), University of Brescia, v.le Europa 11, 25121 Brescia, Italy
| | - Erica Filippi
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Marta Turati
- Molecular and Translational Medicine Dept (DMMT), University of Brescia, v.le Europa 11, 25121 Brescia, Italy
| | - Sofia Parrasia
- Department of Biology (DiBio), University of Padova, Via U. Bassi, 58/B, Padova, Italy
| | - Simone Bernardotto
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Martina Stocco
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Ildikò Szabò
- Department of Biology (DiBio), University of Padova, Via U. Bassi, 58/B, Padova, Italy
| | - Andrea Mattarei
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy
| | - Roberto Ronca
- Molecular and Translational Medicine Dept (DMMT), University of Brescia, v.le Europa 11, 25121 Brescia, Italy.
| | - Margherita Morpurgo
- Pharmaceutical and Pharmacological Sciences Dept (DSF), University of Padova, Via Marzolo, 5. 35131 Padova, Italy.
| |
Collapse
|
2
|
Vasilevich NI, Jiang H, Xiao H, Feng K, Jian C, Chen C, Li M, Chen Z, Pang L, Li X, Chestkov AV, Sun AH, Xu W, Fuselier JA, Coy DH, Sun L. Biological evaluation of 9-thioansamitocin P3. Biochem Biophys Res Commun 2024; 696:149483. [PMID: 38219484 DOI: 10.1016/j.bbrc.2024.149483] [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: 11/08/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Highly cytotoxic maytansine derivatives are widely used in targeted tumor delivery. Structure-activity studies published earlier suggested the C9 carbinol to be a key element necessary to retain the potency. However, in 1984 a patent was published by Takeda in which the synthesis of 9-thioansamitocyn (AP3SH) was described and its activity in xenograft models was shown. In this article we summarize the results of an extended study of the anti-tumor properties of AP3SH. Like other maytansinoids, it induces apoptosis and arrests the cell cycle in the G2/M phase. It is metabolized in liver microsomes predominately by C3A4 isoform and doesn't inhibit any CYP isoforms except CYP3A4 (midazolam, IC50 7.84 μM). No hERG inhibition, CYP induction or mutagenicity in Ames tests were observed. AP3SH demonstrates high antiproliferative activity against 25 tumor cell lines and tumor growth inhibition in U937 xenograft model. Application of AP3SH as a cytotoxic payload in drug delivery system was demonstrated by us earlier.
Collapse
Affiliation(s)
- Natalya I Vasilevich
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China.
| | - Huangyu Jiang
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Haihua Xiao
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Kunxian Feng
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Chengfang Jian
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Changfeng Chen
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Min Li
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Zhenhua Chen
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Li Pang
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Xiang Li
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Alexander V Chestkov
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China
| | - Andre H Sun
- Department of Pathology, Tulane University Health Sciences Center, Tulane Cancer Center, New Orleans, LA70112, USA
| | - Wang Xu
- Department of Pathology, Tulane University Health Sciences Center, Tulane Cancer Center, New Orleans, LA70112, USA
| | - Joseph A Fuselier
- Peptide Research Labs, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - David H Coy
- Peptide Research Labs, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Lichun Sun
- Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd, Shenzhen, Guangdong, 518118, China; Peptide Research Labs, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA.
| |
Collapse
|
3
|
Nguyen TD, Bordeau BM, Balthasar JP. Use of Payload Binding Selectivity Enhancers to Improve Therapeutic Index of Maytansinoid-Antibody-Drug Conjugates. Mol Cancer Ther 2023; 22:1332-1342. [PMID: 37493255 PMCID: PMC10811745 DOI: 10.1158/1535-7163.mct-22-0804] [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: 12/15/2022] [Revised: 05/03/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
Systemic exposure to released cytotoxic payload contributes to the dose-limiting off-target toxicities of anticancer antibody-drug conjugates (ADC). In this work, we present an "inverse targeting" strategy to optimize the therapeutic selectivity of maytansinoid-conjugated ADCs. Several anti-maytansinoid sdAbs were generated via phage-display technology with binding IC50 values between 10 and 60 nmol/L. Co-incubation of DM4 with the anti-maytansinoid sdAbs shifted the IC50 value of DM4 up to 250-fold. Tolerability and efficacy of 7E7-DM4 ADC, an anti-CD123 DM4-conjugated ADC, were assessed in healthy and in tumor-bearing mice, with and without co-administration of an anti-DM4 sdAb. Co-administration with anti-DM4 sdAb reduced 7E7-DM4-induced weight loss, where the mean values of percentage weight loss at nadir for mice receiving ADC+saline and ADC+sdAb were 7.9% ± 3% and 3.8% ± 1.3% (P < 0.05). In tumor-bearing mice, co-administration of the anti-maytansinoid sdAb did not negatively affect the efficacy of 7E7-DM4 on tumor growth or survival following dosing of the ADC at 1 mg/kg (P = 0.49) or at 10 mg/kg (P = 0.9). Administration of 7E7-DM4 at 100 mg/kg led to dramatic weight loss, with 80% of treated mice succumbing to toxicity before the appearance of mortality relating to tumor growth in control mice. However, all mice receiving co-dosing of 100 mg/kg 7E7-DM4 with anti-DM4 sdAb were able to tolerate the treatment, which enabled reduction in tumor volume to undetectable levels and to dramatic improvements in survival. In summary, we have demonstrated the utility and feasibility of the application of anti-payload antibody fragments for inverse targeting to improve the selectivity and efficacy of anticancer ADC therapy.
Collapse
Affiliation(s)
- Toan D. Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214
| | - Brandon M. Bordeau
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214
| | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214
| |
Collapse
|
4
|
Najjar MK, Manore SG, Regua AT, Lo HW. Antibody-Drug Conjugates for the Treatment of HER2-Positive Breast Cancer. Genes (Basel) 2022; 13:2065. [PMID: 36360302 PMCID: PMC9691220 DOI: 10.3390/genes13112065] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) receptor tyrosine kinase is overexpressed in 20-30% of breast cancers and is associated with poor prognosis and worse overall patient survival. Most women with HER2-positive breast cancer receive neoadjuvant chemotherapy plus HER2-targeted therapies. The development of HER2-directed therapeutics is an important advancement in targeting invasive breast cancer. Despite the efficacy of anti-HER2 monoclonal antibodies, they are still being combined with adjuvant chemotherapy to improve overall patient outcomes. Recently, significant progress has been made towards the development of a class of therapeutics known as antibody-drug conjugates (ADCs), which leverage the high specificity of HER2-targeted monoclonal antibodies with the potent cytotoxic effects of various small molecules, such as tubulin inhibitors and topoisomerase inhibitors. To date, two HER2-targeting ADCs have been approved by the FDA for the treatment of HER2-positive breast cancer: Ado-trastuzumab emtansine (T-DM1; Kadcyla®) and fam-trastuzumab deruxtecan-nxki (T-Dxd; Enhertu®). Kadcyla and Enhertu are approved for use as a second-line treatment after trastuzumab-taxane-based therapy in patients with HER2-positive breast cancer. The success of ADCs in the treatment of HER2-positive breast cancer provides novel therapeutic advancements in the management of the disease. In this review, we discuss the basic biology of HER2, its downstream signaling pathways, currently available anti-HER2 therapeutic modalities and their mechanisms of action, and the latest clinical and safety characteristics of ADCs used for the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Mariana K. Najjar
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Sara G. Manore
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Angelina T. Regua
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, MSE R162, 6431 Fannin Street, Houston, TX 77030, USA
| | - Hui-Wen Lo
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, MSE R162, 6431 Fannin Street, Houston, TX 77030, USA
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
5
|
Pouzin C, Tod M, Chadjaa M, Fagniez N, Nguyen L. Covariate analysis of tusamitamab ravtansine, a DM4 anti‐CEACAM5 antibody‐drug conjugate, based on first‐in‐human study. CPT Pharmacometrics Syst Pharmacol 2022; 11:384-394. [PMID: 35191618 PMCID: PMC8923727 DOI: 10.1002/psp4.12769] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/12/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022] Open
Abstract
Tusamitamab ravtansine is an anti‐CEACAM5 antibody‐drug conjugate indicated in patients with solid tumors. Based on a previous developed semimechanistic model describing simultaneously pharmacokinetic (PK) of SAR408701, two of its active metabolites: DM4 and methyl‐DM4 and naked antibody, with integration of drug‐to‐antibody data, the main objective of the present analysis was to evaluate covariate’s impact in patients from phase I/II study (n = 254). Demographic and pathophysiologic baseline covariates were explored to explain interindividual variability on each entity PK parameter. Model parameters were estimated with good precision. Five covariates were included in the final PK model: body surface area (BSA), tumor burden, albumin, circulating target, and gender. Comparison of BSA‐adjusted dosing and flat dosing supported the current BSA‐based dosing regimen, to limit under and over exposure in patients with extreme BSA. Overall, this model characterized accurately the PKs of all entities and highlighted sources of PK variability. By integrating mechanistic considerations, this model aimed to improve understanding of the SAR408701 complex disposition while supporting key steps of clinical development.
Collapse
Affiliation(s)
- Clemence Pouzin
- Pharmacokinetics Dynamics and Metabolism Department Sanofi R&D Paris France
- PKPD Modelling Unit Oncology Department EMR3738 University of Claude Bernard Lyon 1 Lyon France
| | - Michel Tod
- PKPD Modelling Unit Oncology Department EMR3738 University of Claude Bernard Lyon 1 Lyon France
| | | | - Nathalie Fagniez
- Pharmacokinetics Dynamics and Metabolism Department Sanofi R&D Paris France
| | - Laurent Nguyen
- Pharmacokinetics Dynamics and Metabolism Department Sanofi R&D Paris France
| |
Collapse
|
6
|
Pouzin C, Gibiansky L, Fagniez N, Chadjaa M, Tod M, Nguyen L. Integrated multiple analytes and semi-mechanistic population pharmacokinetic model of tusamitamab ravtansine, a DM4 anti-CEACAM5 antibody-drug conjugate. J Pharmacokinet Pharmacodyn 2022; 49:381-394. [PMID: 35166967 PMCID: PMC9098589 DOI: 10.1007/s10928-021-09799-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/20/2021] [Indexed: 01/01/2023]
Abstract
Tusamitamab ravtansine (SAR408701) is an antibody-drug conjugate (ADC), combining a humanized monoclonal antibody (IgG1) targeting carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) and a potent cytotoxic maytansinoid derivative, DM4, inhibiting microtubule assembly. SAR408701 is currently in clinical development for the treatment of advanced solid tumors expressing CEACAM5. It is administered intravenously as a conjugated antibody with an average Drug Antibody Ratio (DAR) of 3.8. During SAR408701 clinical development, four entities were measured in plasma: conjugated antibody (SAR408701), naked antibody (NAB), DM4 and its methylated metabolite (MeDM4), both being active. Average DAR and proportions of individual DAR species were also assessed in a subset of patients. An integrated and semi-mechanistic population pharmacokinetic model describing the time-course of all entities in plasma and DAR measurements has been developed. All DAR moieties were assumed to share the same drug disposition parameters, excepted for clearance which differed for DAR0 (i.e. NAB entity). The conversion of higher DAR to lower DAR resulted in a DAR-dependent ADC deconjugation and was represented as an irreversible first-order process. Each conjugated antibody was assumed to contribute to DM4 formation. All data were fitted simultaneously and the model developed was successful in describing the pharmacokinetic profile of each entity. Such a structural model could be translated to other ADCs and gives insight of mechanistic processes governing ADC disposition. This framework will further be expanded to evaluate covariates impact on SAR408701 pharmacokinetics and its derivatives, and thus can help identifying sources of pharmacokinetic variability and potential efficacy and safety pharmacokinetic drivers.
Collapse
Affiliation(s)
- Clemence Pouzin
- Sanofi R&D, Pharmacokinetics Dynamics and Metabolism Department, 1 Avenue Pierre Brossolette, Chilly-Mazarin, 91380, Paris, France.
- Oncology department EMR3738, PKPD modelling unit, University of Claude Bernard Lyon 1, Lyon, France.
| | | | - Nathalie Fagniez
- Sanofi R&D, Pharmacokinetics Dynamics and Metabolism Department, 1 Avenue Pierre Brossolette, Chilly-Mazarin, 91380, Paris, France
| | | | - Michel Tod
- Oncology department EMR3738, PKPD modelling unit, University of Claude Bernard Lyon 1, Lyon, France
| | - Laurent Nguyen
- Sanofi R&D, Pharmacokinetics Dynamics and Metabolism Department, 1 Avenue Pierre Brossolette, Chilly-Mazarin, 91380, Paris, France
| |
Collapse
|
7
|
Usama SM, Marker SC, Caldwell DR, Patel NL, Feng Y, Kalen JD, St Croix B, Schnermann MJ. Targeted Fluorogenic Cyanine Carbamates Enable In Vivo Analysis of Antibody-Drug Conjugate Linker Chemistry. J Am Chem Soc 2021; 143:21667-21675. [PMID: 34928588 DOI: 10.1021/jacs.1c10482] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibody-drug conjugates (ADCs) are a rapidly emerging therapeutic platform. The chemical linker between the antibody and the drug payload plays an essential role in the efficacy and tolerability of these agents. New methods that quantitatively assess the cleavage efficiency in complex tissue settings could provide valuable insights into the ADC design process. Here we report the development of a near-infrared (NIR) optical imaging approach that measures the site and extent of linker cleavage in mouse models. This approach is enabled by a superior variant of our recently devised cyanine carbamate (CyBam) platform. We identify a novel tertiary amine-containing norcyanine, the product of CyBam cleavage, that exhibits a dramatically increased cellular signal due to an improved cellular permeability and lysosomal accumulation. The resulting cyanine lysosome-targeting carbamates (CyLBams) are ∼50× brighter in cells, and we find this strategy is essential for high-contrast in vivo targeted imaging. Finally, we compare a panel of several common ADC linkers across two antibodies and tumor models. These studies indicate that cathepsin-cleavable linkers provide dramatically higher tumor activation relative to hindered or nonhindered disulfides, an observation that is only apparent with in vivo imaging. This strategy enables quantitative comparisons of cleavable linker chemistries in complex tissue settings with implications across the drug delivery landscape.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Sierra C Marker
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Donald R Caldwell
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Nimit L Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland 21702, United States
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, Maryland 21702, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
8
|
Khera E, Cilliers C, Smith MD, Ganno ML, Lai KC, Keating TA, Kopp A, Nessler I, Abu-Yousif AO, Thurber GM. Quantifying ADC bystander payload penetration with cellular resolution using pharmacodynamic mapping. Neoplasia 2020; 23:210-221. [PMID: 33385970 PMCID: PMC7779838 DOI: 10.1016/j.neo.2020.12.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
With the recent approval of 3 new antibody drug conjugates (ADCs) for solid tumors, this class of drugs is gaining momentum for the targeted treatment of cancer. Despite significant investment, there are still fundamental issues that are incompletely understood. Three of the recently approved ADCs contain payloads exhibiting bystander effects, where the payload can diffuse out of a targeted cell into adjacent cells. These effects are often studied using a mosaic of antigen positive and negative cells. However, the distance these payloads can diffuse in tumor tissue while maintaining a lethal concentration is unclear. Computational studies suggest bystander effects partially compensate for ADC heterogeneity in tumors in addition to targeting antigen negative cells. However, this type of study is challenging to conduct experimentally due to the low concentrations of extremely potent payloads. In this work, we use a series of 3-dimensional cell culture and primary human tumor xenograft studies to directly track fluorescently labeled ADCs and indirectly follow the payload via an established pharmacodynamic marker (γH2A. X). Using TAK-164, an anti-GCC ADC undergoing clinical evaluation, we show that the lipophilic DNA-alkylating payload, DGN549, penetrates beyond the cell targeted layer in GCC-positive tumor spheroids and primary human tumor xenograft models. The penetration distance is similar to model predictions, where the lipophilicity results in moderate tissue penetration, thereby balancing improved tissue penetration with sufficient cellular uptake to avoid significant washout. These results aid in mechanistic understanding of the interplay between antigen heterogeneity, bystander effects, and heterogeneous delivery of ADCs in the tumor microenvironment to design clinically effective therapeutics.
Collapse
Affiliation(s)
- Eshita Khera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Cornelius Cilliers
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | | | - Anna Kopp
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Ian Nessler
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Lazzerini L, Jöhrens K, Sehouli J, Cichon G. Favorable therapeutic response after anti-Mesothelin antibody-drug conjugate treatment requires high expression of Mesothelin in tumor cells. Arch Gynecol Obstet 2020; 302:1255-1262. [PMID: 32815024 PMCID: PMC7524828 DOI: 10.1007/s00404-020-05734-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 07/31/2020] [Indexed: 11/29/2022]
Abstract
PURPOSE The cell surface glycoprotein Mesothelin is overexpressed in ovarian, fallopian tube, endometrial, cervical and primary peritoneal cancer and, therefore, might become a particular interesting tumor target in gynecologic oncology. However, even in malignant tumors of the same entity the level of Mesothelin expression varies between individuals, hence it can be expected that the response to Mesothelin-targeting therapies will be variable as well. In this study we explored the therapeutic potency of a novel anti-Mesothelin antibody-drug conjugate (Anetumab ravtansine) as a function of Mesothelin expression in the targeted tumor cells. METHODS Anti-tumor activity studies were performed in human uterine xenograft tumor models that express Mesothelin at high, moderate or low levels. The antibody-drug conjugate (ADC) was applied in varying doses ranging from 2 to 15 mg/kg at variable intervals in tumor bearing SCID/beige mice and the impact on tumor growth was monitored. RESULTS The therapeutic response to the anti-Mesothelin ADC correlated closely with the level of Mesothelin expression in tumor cells. Within the applied dose levels complete tumor regression was achieved only in tumors which expressed Mesothelin at particularly high levels (Hela cell tumors). The application of high anti-Mesothelin ADC doses less frequently was more efficious than giving lower doses at a higher frequency. CONCLUSION The studies confirm the great therapeutic potential of Anetumab ravtansine. However, a favorable treatment outcome requires strong Mesothelin expression in tumor cells. Future clinical trials may benefit from a more rigorous selection of appropriate patients based on the level of Mesothelin expression in their tumor tissue. If, in addition, it is possible to better control side effects by introducing protective measures and by doing so to increase the maximum tolerated dose, Anetumab ravtansine has the potency to become a valuable therapeutic tool, especially in the field of gynecological oncology.
Collapse
Affiliation(s)
- Lea Lazzerini
- Department of Gynecology, Charité, Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Korinna Jöhrens
- Department of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, Schubertstrasse 15, 01307, Dresden, Germany
| | - Jalid Sehouli
- Department of Gynecology, Charité, Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Günter Cichon
- Department of Gynecology, Charité, Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Germany.
| |
Collapse
|
10
|
Zuo P. Capturing the Magic Bullet: Pharmacokinetic Principles and Modeling of Antibody-Drug Conjugates. AAPS JOURNAL 2020; 22:105. [PMID: 32767003 DOI: 10.1208/s12248-020-00475-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
Abstract
Over the past two decades, antibody-drug conjugates (ADCs) have emerged as a promising class of drugs for cancer therapy and have expanded to nononcology fields such as inflammatory diseases, atherosclerosis, and bacteremia. Eight ADCs are currently approved by FDA for clinical applications, with more novel ADCs under clinical development. Compared with traditional chemotherapy, ADCs combine the target specificity of antibodies with chemotherapeutic capabilities of cytotoxic drugs. The benefits include reduced systemic toxicity and enhanced therapeutic index for patients. However, the heterogeneous structures of ADCs and their dynamic changes following administration create challenges in their development. The understanding of ADC pharmacokinetics (PK) is crucial for the optimization of clinical dosing regimens when translating from animal to human. In addition, it contributes to the optimization of dose selection and clinical monitoring with regard to safety and efficacy. This manuscript reviews the PK characteristics of ADCs and summarizes the diverse approaches for PK modeling that can be used to evaluate an ADC at the preclinical and clinical stages to support their successful development. Despite the numerous available options, fit-for-purpose modeling approaches for the PK and PD of ADCs should be critically planned and well-thought-out to adequately support the development of an ADC.
Collapse
Affiliation(s)
- Peiying Zuo
- Pharmacometrics US, Clinical Pharmacology & Exploratory Development, Astellas Pharma, Inc., USA, 1 Astellas Way, Northbrook, Illinois, 60062, USA.
| |
Collapse
|
11
|
Falgàs A, Pallarès V, Serna N, Sánchez-García L, Sierra J, Gallardo A, Alba-Castellón L, Álamo P, Unzueta U, Villaverde A, Vázquez E, Mangues R, Casanova I. Selective delivery of T22-PE24-H6 to CXCR4 + diffuse large B-cell lymphoma cells leads to wide therapeutic index in a disseminated mouse model. Theranostics 2020; 10:5169-5180. [PMID: 32373205 PMCID: PMC7196303 DOI: 10.7150/thno.43231] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Novel therapeutic strategies are urgently needed to reduce relapse rates and enhance survival in Diffuse Large B-Cell Lymphoma (DLBCL) patients. CXCR4-overexpressing cancer cells are good targets for therapy because of their association with dissemination and relapse in R-CHOP treated DLBCL patients. Immunotoxins that incorporate bacterial toxins are potentially effective in treating haematological neoplasias, but show a narrow therapeutic index due to the induction of severe side effects. Therefore, when considering the delivery of these toxins as cancer therapeutics, there is a need not only to increase their uptake in the target cancer cells, and their stability in blood, but also to reduce their systemic toxicity. We have developed a therapeutic nanostructured protein T22-PE24-H6 that incorporates exotoxin A from Pseudomonas aeruginosa, which selectively targets lymphoma cells because of its specific interaction with a highly overexpressed CXCR4 receptor (CXCR4+) in DLBCL. Methods: T22-PE24-H6 cytotoxicity and its dependence on the CXCR4 receptor were evaluated in DLBCL cell lines using cell viability assays. Different in vitro experiments (mitochondrial membrane potential, Western Blot, Annexin V and DAPI staining) were conducted to determine T22-PE24-H6 cell death mechanisms. In vivo imaging and therapeutic effect studies were performed in a disseminated DLBCL mouse model that mimics organ infiltration in DLBCL patients. Finally, immunohistochemistry and histopathology analyses were used to evaluate the antineoplastic effect and systemic toxicity. Results: In vitro, T22-PE24-H6 induced selective cell death of CXCR4+ DLBCL cells by activating the apoptotic pathway. In addition, repeated T22-PE24-H6 intravenous administration in a CXCR4+ DLBCL-disseminated mouse model showed a significant reduction of lymphoma burden in organs clinically affected by DLBCL cells (lymph nodes and bone marrow). Finally, we did not observe systemic toxicity associated to the nanoparticle treatment in non-DLBCL-infiltrated organs. Conclusion: We have demonstrated here a potent T22-PE24-H6 antineoplastic effect, especially in blocking dissemination in a CXCR4+ DLBCL model without associated toxicity. Thereby, T22-PE24-H6 promises to become an effective alternative to treat CXCR4+ disseminated refractory or relapsed DLBCL patients.
Collapse
|
12
|
Mertansine Inhibits mRNA Expression and Enzyme Activities of Cytochrome P450s and Uridine 5′-Diphospho-Glucuronosyltransferases in Human Hepatocytes and Liver Microsomes. Pharmaceutics 2020; 12:pharmaceutics12030220. [PMID: 32131538 PMCID: PMC7150891 DOI: 10.3390/pharmaceutics12030220] [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: 02/18/2020] [Revised: 02/28/2020] [Accepted: 03/01/2020] [Indexed: 11/16/2022] Open
Abstract
Mertansine, a tubulin inhibitor, is used as the cytotoxic component of antibody–drug conjugates (ADCs) for cancer therapy. The effects of mertansine on uridine 5′-diphospho-glucuronosyltransferase (UGT) activities in human liver microsomes and its effects on the mRNA expression of cytochrome P450s (CYPs) and UGTs in human hepatocytes were evaluated to assess the potential for drug–drug interactions (DDIs). Mertansine potently inhibited UGT1A1-catalyzed SN-38 glucuronidation, UGT1A3-catalyzed chenodeoxycholic acid 24-acyl-β-glucuronidation, and UGT1A4-catalyzed trifluoperazine N-β-d-glucuronidation, with Ki values of 13.5 µM, 4.3 µM, and 21.2 µM, respectively, but no inhibition of UGT1A6, UGT1A9, and UGT2B7 enzyme activities was observed in human liver microsomes. A 48 h treatment of mertansine (1.25–2500 nM) in human hepatocytes resulted in the dose-dependent suppression of mRNA levels of CYP1A2, CYP2B6, CYP3A4, CYP2C8, CYP2C9, CYP2C19, UGT1A1, and UGT1A9, with IC50 values of 93.7 ± 109.1, 36.8 ± 18.3, 160.6 ± 167.4, 32.1 ± 14.9, 578.4 ± 452.0, 539.5 ± 233.4, 856.7 ± 781.9, and 54.1 ± 29.1 nM, respectively, and decreased the activities of CYP1A2-mediated phenacetin O-deethylase, CYP2B6-mediated bupropion hydroxylase, and CYP3A4-mediated midazolam 1′-hydroxylase. These in vitro DDI potentials of mertansine with CYP1A2, CYP2B6, CYP2C8/9/19, CYP3A4, UGT1A1, and UGT1A9 substrates suggest that it is necessary to carefully characterize the DDI potentials of ADC candidates with mertansine as a payload in the clinic.
Collapse
|
13
|
Liu H, Bolleddula J, Nichols A, Tang L, Zhao Z, Prakash C. Metabolism of bioconjugate therapeutics: why, when, and how? Drug Metab Rev 2020; 52:66-124. [PMID: 32045530 DOI: 10.1080/03602532.2020.1716784] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bioconjugation of therapeutic agents has been used as a selective drug delivery platform for many therapeutic areas. Bioconjugates are prepared by the covalent linkage of active compounds (small or large molecule) to a carrier molecule (lipids, proteins, peptides, carbohydrates, and polymers) through a chemical linker. The linkage of the active component to a carrier molecule enhances the therapeutic window through a targeted delivery and by reducing toxicity. Bioconjugates also possess improved pharmacokinetic properties such as a long half-life, increased stability, and cleavage by intracellular enzymes/environment. However, premature cleavage of the bioconjugates and the resulting metabolites/catabolites may produce undesirable toxic effects and, hence, it is critical to understand cleavage mechanisms, metabolism of bioconjugates, and translatability to human in the discovery stages. This article provides a comprehensive overview of linker cleavage pathways and catabolism/metabolism of antibody-drug conjugates, glycoconjugates, polymer-drug conjugates, lipid-drug conjugates, folate-targeted small molecule-drug conjugates, and drug-drug conjugates.
Collapse
Affiliation(s)
- Hanlan Liu
- KSQ Therapeutics Inc., Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
14
|
Costoplus JA, Veale KH, Qiu Q, Ponte JF, Lanieri L, Setiady Y, Dong L, Skaletskaya A, Bartle LM, Salomon P, Wu R, Maloney EK, Kovtun YV, Ab O, Lai K, Chari RVJ, Widdison WC. Peptide-Cleavable Self-immolative Maytansinoid Antibody-Drug Conjugates Designed To Provide Improved Bystander Killing. ACS Med Chem Lett 2019; 10:1393-1399. [PMID: 31620224 PMCID: PMC6792174 DOI: 10.1021/acsmedchemlett.9b00310] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/27/2019] [Indexed: 11/29/2022] Open
Abstract
![]()
A new type of antibody–drug
conjugate (ADC) has been prepared that contains a sulfur-bearing maytansinoid
attached to an antibody via a highly stable tripeptide linker. Once
internalized by cells, proteases in catabolic vesicles cleave the
peptide of the ADC’s linker causing self-immolation that releases
a thiol-bearing metabolite, which is then S-methylated.
Conjugates were prepared with peptide linkers containing only alanyl
residues, which were all l isomers or had a single d residue in one of the three positions. A d-alanyl residue
in the linker did not significantly impair a conjugate’s cytotoxicity
or bystander killing unless it was directly attached to the immolative
moiety. Increasing the number of methylene units in the maytansinoid
side chain of a conjugate did not typically affect an ADC’s
cytotoxicity to targeted cells but did increase bystander killing
activity. ADCs with the highest in vitro bystander
killing were then evaluated in vivo in mice, where
they displayed improved efficacy compared to previously described
types of maytansinoid conjugates.
Collapse
Affiliation(s)
- Juliet A. Costoplus
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Karen H. Veale
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Qifeng Qiu
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Jose F. Ponte
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Leanne Lanieri
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yulius Setiady
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ling Dong
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Anna Skaletskaya
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Laura M. Bartle
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Paulin Salomon
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Rui Wu
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Erin K. Maloney
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yelena V. Kovtun
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Olga Ab
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Kate Lai
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ravi V. J. Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Wayne C. Widdison
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
15
|
Li W, Veale KH, Qiu Q, Sinkevicius KW, Maloney EK, Costoplus JA, Lau J, Evans HL, Setiady Y, Ab O, Abbott SM, Lee J, Wisitpitthaya S, Skaletskaya A, Wang L, Keating TA, Chari RVJ, Widdison WC. Synthesis and Evaluation of Camptothecin Antibody-Drug Conjugates. ACS Med Chem Lett 2019; 10:1386-1392. [PMID: 31620223 PMCID: PMC6792168 DOI: 10.1021/acsmedchemlett.9b00301] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/06/2019] [Indexed: 01/23/2023] Open
Abstract
Antibody-drug conjugates (ADCs) that incorporate the exatecan derivative DXd in their payload are showing promising clinical results in solid tumor indications. The payload has an F-ring that also contains a second chiral center, both of which complicate its synthesis and derivatization. Here we report on new camptothecin-ADCs that do not have an F-ring in their payloads yet behave similarly to DXd-bearing conjugates in vitro and in vivo. This simplification allows easier derivatization of camptothecin A and B rings for structure-activity relationship studies and payload optimization. ADCs having different degrees of bystander killing and the ability to release hydroxyl or thiol-bearing metabolites following peptide linker cleavage were investigated.
Collapse
Affiliation(s)
- Wei Li
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Karen H. Veale
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Qifeng Qiu
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | | | - Erin K. Maloney
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Juliet A. Costoplus
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Janet Lau
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Helen L. Evans
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yulius Setiady
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Olga Ab
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Stephen M. Abbott
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Jenny Lee
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | | | - Anna Skaletskaya
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Lintao Wang
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Thomas A. Keating
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ravi V. J. Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Wayne C. Widdison
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
16
|
Yoder NC, Bai C, Tavares D, Widdison WC, Whiteman KR, Wilhelm A, Wilhelm SD, McShea MA, Maloney EK, Ab O, Wang L, Jin S, Erickson HK, Keating TA, Lambert JM. A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates the Benefits of Lysine Conjugation. Mol Pharm 2019; 16:3926-3937. [PMID: 31287952 DOI: 10.1021/acs.molpharmaceut.9b00529] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates are an emerging class of cancer therapeutics constructed from monoclonal antibodies conjugated with small molecule effectors. First-generation molecules of this class often employed heterogeneous conjugation chemistry, but many site-specifically conjugated ADCs have been described recently. Here, we undertake a systematic comparison of ADCs made with the same antibody and the same macrocyclic maytansinoid effector but conjugated either heterogeneously at lysine residues or site-specifically at cysteine residues. Characterization of these ADCs in vitro reveals generally similar properties, including a similar catabolite profile, a key element in making a meaningful comparison of conjugation chemistries. In a mouse model of cervical cancer, the lysine-conjugated ADC affords greater efficacy on a molar payload basis. Rather than making general conclusions about ADCs conjugated by a particular chemistry, we interpret these results as highlighting the complexity of ADCs and the interplay between payload class, linker chemistry, target antigen, and other variables that determine efficacy in a given setting.
Collapse
|
17
|
Zheng Y, Shen Y, Meng X, Wu Y, Zhao Y, Wu C. Stabilizing p-Dithiobenzyl Urethane Linkers without Rate-Limiting Self-Immolation for Traceless Drug Release. ChemMedChem 2019; 14:1196-1203. [PMID: 31020782 DOI: 10.1002/cmdc.201900248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Indexed: 12/31/2022]
Abstract
Exploiting the redox sensitivity of disulfide bonds is a prevalent strategy in targeted prodrug designs. In contrast to aliphatic disulfides, p-thiobenzyl-based disulfides have rarely been used for prodrug designs, given their intrinsic instability caused by the low pKa of aromatic thiols. Here, we examined the interplay between steric hindrance and the low-pKa effect on thiol-disulfide exchange reactions and uncovered a new thiol-disulfide exchange process for the self-immolation of p-thiobenzyl-based disulfides. We observed a central leaving group shifting effect in the α,α-dimethyl-substituted p-dithiobenzyl urethane linkers (DMTB linkers), which leads to increased disulfide stability by more than two orders of magnitude, an extent that is significantly greater than that observed with typical aliphatic disulfides. In particular, the DMTB linkers display not only high stability, but also rapid self-immolation kinetics due to the low pKa of the aromatic thiol, which can be used as a general and robust linkage between targeting reagents and cytotoxic drugs for targeted prodrug designs. The unique and promising stability characteristics of the present DMTB linker will likely inspire the development of novel targeted prodrugs to achieve traceless release of drugs into cells.
Collapse
Affiliation(s)
- Yiwu Zheng
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Yang Shen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Xiaoting Meng
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Yaqi Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Yibing Zhao
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Chuanliu Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| |
Collapse
|
18
|
Lepelley M, Allouchery M, Long J, Boucherle D, Ranchoup Y, Le Marc'Hadour F, Villier C, Sturm N. Nodular Regenerative Hyperplasia Induced by Trastuzumab Emtansine: Role of Emtansine? Ann Hepatol 2018; 17:1067-1071. [PMID: 30600283 DOI: 10.5604/01.3001.0012.7207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Trastuzumab is a monoclonal antibody targeted against the Human Epidermal Growth Factor Receptor 2 (HER2) overexpressed in some breast cancer. This targeted therapy significantly improves the prognosis of these cancers. Recently an anti-HER2 antibodydrug conjugate was shaped in order to facilitate the targeted delivery of potent cytotoxic drug to cancer cells and to reduce resistance. This formulation, called trastuzumab emtansine (T-DM1), consists of the monoclonal antibody trastuzumab linked to a cytotoxic drug (a derivative of maytansine) via a chemical linker. Little is known about adverse reactions due to this new formulation. Herein we described the case of a woman suffering from a HER2-positive breast cancer, treated with trastuzumab for 30 months followed by T-DM1 monotherapy. After 12 months of T-DM1 treatment, a nodular regenerative hyperplasia confirmed by liver biopsy occurred. T-DM1 was stopped and medical imagery showed a resolution of the nodular regenerative hyperplasia. Unfortunately, hepatic metastasis progressed. Few cases of nodular regenerative hyperplasia induced by T-DM1 have been described so far. Further studies are needed to explore pathogenesis of nodular regenerative hyperplasia with this new antibody-drug conjugate treatment.
Collapse
Affiliation(s)
- Marion Lepelley
- Centre régional de pharmacovigilance, CHU de Grenoble-Alpes, Grenoble, France. † Pharmacy, Groupe Hospitalier Mutualiste, Grenoble, France
| | | | - Jérôme Long
- Oncology, Groupe Hospitalier Mutualiste, Grenoble, France
| | | | | | | | - Céline Villier
- Centre régional de pharmacovigilance, CHU de Grenoble-Alpes, Grenoble, France
| | - Nathalie Sturm
- Department of pathology, CHU de Grenoble-Alpes, Grenoble, France
| |
Collapse
|
19
|
Karpov AS, Abrams T, Clark S, Raikar A, D’Alessio JA, Dillon MP, Gesner TG, Jones D, Lacaud M, Mallet W, Martyniuk P, Meredith E, Mohseni M, Nieto-Oberhuber CM, Palacios D, Perruccio F, Piizzi G, Zurini M, Bialucha CU. Nicotinamide Phosphoribosyltransferase Inhibitor as a Novel Payload for Antibody-Drug Conjugates. ACS Med Chem Lett 2018; 9:838-842. [PMID: 30128077 PMCID: PMC6088352 DOI: 10.1021/acsmedchemlett.8b00254] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 06/28/2018] [Indexed: 11/28/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a novel modality that allows targeted delivery of potent therapeutic agents to the desired site. Herein we report our discovery of NAMPT inhibitors as a novel nonantimitotic payload for ADCs. The resulting anti-c-Kit conjugates (ADC-3 and ADC-4) demonstrated in vivo efficacy in the c-Kit positive gastrointestinal stromal tumor GIST-T1 xenograft model in a target-dependent manner.
Collapse
Affiliation(s)
- Alexei S. Karpov
- Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Tinya Abrams
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Suzanna Clark
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Ankita Raikar
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Joseph A. D’Alessio
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Michael P. Dillon
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Thomas G. Gesner
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Darryl Jones
- Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Marion Lacaud
- Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - William Mallet
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Piotr Martyniuk
- Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Erik Meredith
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Morvarid Mohseni
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | | | - Daniel Palacios
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | | | - Grazia Piizzi
- Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Mauro Zurini
- Novartis Institutes for BioMedical Research, CH-4056 Basel, Switzerland
| | - Carl Uli Bialucha
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
20
|
Duan B, Wang D, Wu H, Xu P, Jiang P, Xia G, Liu Z, Wang H, Guo Z, Chen Q. Core–Shell Structurized Fe3O4@C@MnO2 Nanoparticles as pH Responsive T1-T2* Dual-Modal Contrast Agents for Tumor Diagnosis. ACS Biomater Sci Eng 2018; 4:3047-3054. [DOI: 10.1021/acsbiomaterials.8b00287] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Beichen Duan
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science and Engineering, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230026, People’s Republic of China
| | - Dongdong Wang
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science and Engineering, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230026, People’s Republic of China
| | - Huihui Wu
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230027, People’s Republic of China
| | - Pengping Xu
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science and Engineering, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230026, People’s Republic of China
| | - Peng Jiang
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science and Engineering, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230026, People’s Republic of China
| | - Guoliang Xia
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science and Engineering, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230026, People’s Republic of China
| | - Zhenbang Liu
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230027, People’s Republic of China
| | - Haibao Wang
- Radiology Department of the First Affiliated Hospital of Anhui Medical University, No.218, Jixi Road, Hefei, 230022, People’s Republic of China
| | - Zhen Guo
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230027, People’s Republic of China
| | - Qianwang Chen
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science and Engineering, University of Science and Technology of China, No.96, JinZhai Road, Hefei 230026, People’s Republic of China
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, No.350 Shushanhu Road, Hefei 230031, People’s Republic of China
| |
Collapse
|
21
|
Snyder JT, Malinao MC, Dugal-Tessier J, Atkinson JE, Anand BS, Okada A, Mendelsohn BA. Metabolism of an Oxime-Linked Antibody Drug Conjugate, AGS62P1, and Characterization of Its Identified Metabolite. Mol Pharm 2018; 15:2384-2390. [PMID: 29757653 DOI: 10.1021/acs.molpharmaceut.8b00225] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AGS62P1 is an antibody drug conjugate (ADC) composed of a human IgG1κ monoclonal antibody against FLT3 (FMS-like tyrosine kinase 3) with a p-acetyl phenylalanine (pAF) residue inserted at position 124 of each heavy chain linked to the proprietary microtubule disrupting agent AGL-0182-30 via an alkoxyamine linker that forms an oxime upon conjugation to the antibody. AGS62P1 is currently in Phase I human clinical trials for acute myelogenous leukemia (AML). The identified primary metabolite of an oxime-linked ADC is presented for the first time. AGS62P1 metabolism was assessed in xenograft tumor-bearing mice and rats treated with the ADC using liquid chromatography and mass spectrometry-based methods described herein. In this study, we identified the metabolite of AGS62P1 as pAF-AGL-0185-30, which contains a fragment resulting from the catabolism of the antibody component of the ADC and hydrolysis of the terminal amide portion of the linker-payload. We demonstrated that the metabolite of AGS62P1 is tolerated in rats above 1.5 mg/kg and above 0.334 mg/kg in cynomolgus monkeys when given as a single dose. Furthermore, we established in vitro that pAF-AGL-0185-30 does not significantly inhibit hERG or cytochrome P450 family enzymes (CYPs).
Collapse
Affiliation(s)
- Josh T Snyder
- Agensys Inc. an Affiliate of Astellas Pharma Inc. , 1800 Stewart Street , Santa Monica , California 90404 , United States
| | - Maria-Christina Malinao
- Agensys Inc. an Affiliate of Astellas Pharma Inc. , 1800 Stewart Street , Santa Monica , California 90404 , United States
| | - Julien Dugal-Tessier
- Agensys Inc. an Affiliate of Astellas Pharma Inc. , 1800 Stewart Street , Santa Monica , California 90404 , United States
| | - John E Atkinson
- Agensys Inc. an Affiliate of Astellas Pharma Inc. , 1800 Stewart Street , Santa Monica , California 90404 , United States
| | - Banmeet S Anand
- Agensys Inc. an Affiliate of Astellas Pharma Inc. , 1800 Stewart Street , Santa Monica , California 90404 , United States
| | - Akihiro Okada
- Agensys Inc. an Affiliate of Astellas Pharma Inc. , 1800 Stewart Street , Santa Monica , California 90404 , United States
| | - Brian A Mendelsohn
- Agensys Inc. an Affiliate of Astellas Pharma Inc. , 1800 Stewart Street , Santa Monica , California 90404 , United States
| |
Collapse
|
22
|
Taplin S, Vashisht K, Walles M, Calise D, Kluwe W, Bouchard P, Johnson R. Hepatotoxicity with antibody maytansinoid conjugates: A review of preclinical and clinical findings. J Appl Toxicol 2018; 38:600-615. [DOI: 10.1002/jat.3582] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Sarah Taplin
- Novartis Pharmaceuticals Inc.; East Hanover NJ USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Tubulin Inhibitor-Based Antibody-Drug Conjugates for Cancer Therapy. Molecules 2017; 22:molecules22081281. [PMID: 28763044 PMCID: PMC6152078 DOI: 10.3390/molecules22081281] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/29/2017] [Indexed: 11/16/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a class of highly potent biopharmaceutical drugs generated by conjugating cytotoxic drugs with specific monoclonal antibodies through appropriate linkers. Specific antibodies used to guide potent warheads to tumor tissues can effectively reduce undesired side effects of the cytotoxic drugs. An in-depth understanding of antibodies, linkers, conjugation strategies, cytotoxic drugs, and their molecular targets has led to the successful development of several approved ADCs. These ADCs are powerful therapeutics for cancer treatment, enabling wider therapeutic windows, improved pharmacokinetic/pharmacodynamic properties, and enhanced efficacy. Since tubulin inhibitors are one of the most successful cytotoxic drugs in the ADC armamentarium, this review focuses on the progress in tubulin inhibitor-based ADCs, as well as lessons learned from the unsuccessful ADCs containing tubulin inhibitors. This review should be helpful to facilitate future development of new generations of tubulin inhibitor-based ADCs for cancer therapy.
Collapse
|
24
|
Sadowsky JD, Pillow TH, Chen J, Fan F, He C, Wang Y, Yan G, Yao H, Xu Z, Martin S, Zhang D, Chu P, dela Cruz-Chuh J, O’Donohue A, Li G, Del Rosario G, He J, Liu L, Ng C, Su D, Lewis Phillips GD, Kozak KR, Yu SF, Xu K, Leipold D, Wai J. Development of Efficient Chemistry to Generate Site-Specific Disulfide-Linked Protein– and Peptide–Payload Conjugates: Application to THIOMAB Antibody–Drug Conjugates. Bioconjug Chem 2017. [DOI: 10.1021/acs.bioconjchem.7b00258] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Jack D. Sadowsky
- 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
| | - Jinhua Chen
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Fang Fan
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Changrong He
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Yanli Wang
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Gang Yan
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Hui Yao
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Zijin Xu
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| | - Shanique Martin
- 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
| | - Phillip Chu
- 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
| | - Guangmin Li
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Geoffrey Del Rosario
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jintang He
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Luna Liu
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Carl Ng
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dian Su
- 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
| | - Shang-Fan Yu
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Keyang Xu
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Douglas Leipold
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John Wai
- WuXi AppTec Co., Ltd, 288
Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, PR China
| |
Collapse
|
25
|
LC-MS/MS method for the simultaneous determination of Lys-MCC-DM1, MCC-DM1 and DM1 as potential intracellular catabolites of the antibody-drug conjugate trastuzumab emtansine (T-DM1). J Pharm Biomed Anal 2017; 137:170-177. [PMID: 28131055 DOI: 10.1016/j.jpba.2017.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 01/14/2023]
Abstract
Lysine-MCC-DM1, MCC-DM1 and DM1 are potential catabolites of trastuzumab emtansine (T-DM1). A convenient liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated to detect these catabolites simultaneously in in vitro investigations for the first time. Protein precipitation was utilized to prepare the samples. Chromatographic separation was achieved on a Phenomenex Kinetex C18 column (100×2.1mm, 2.6μm) with mobile-phase gradient elution. The calibration curves of each analyte ranging from 1 to 100nM showed good linearity (r2>0.995). The method was validated successfully and applied to the intracellular catabolism and regulation of T-DM1.
Collapse
|
26
|
Antibody-drug conjugates: Current status and future perspectives. Pharmacol Ther 2016; 167:48-59. [DOI: 10.1016/j.pharmthera.2016.07.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2016] [Indexed: 02/02/2023]
|
27
|
Ogitani Y, Hagihara K, Oitate M, Naito H, Agatsuma T. Bystander killing effect of DS-8201a, a novel anti-human epidermal growth factor receptor 2 antibody-drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity. Cancer Sci 2016; 107:1039-46. [PMID: 27166974 PMCID: PMC4946713 DOI: 10.1111/cas.12966] [Citation(s) in RCA: 416] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/02/2016] [Accepted: 05/06/2016] [Indexed: 12/16/2022] Open
Abstract
Antibody–drug conjugates deliver anticancer agents selectively and efficiently to tumor tissue and have significant antitumor efficacy with a wide therapeutic window. DS‐8201a is a human epidermal growth factor receptor 2 (HER2)‐targeting antibody–drug conjugate prepared using a novel linker‐payload system with a potent topoisomerase I inhibitor, exatecan derivative (DX‐8951 derivative, DXd). It was effective against trastuzumab emtansine (T‐DM1)‐insensitive patient‐derived xenograft models with both high and low HER2 expression. In this study, the bystander killing effect of DS‐8201a was evaluated and compared with that of T‐DM1. We confirmed that the payload of DS‐8201a, DXd (1), was highly membrane‐permeable whereas that of T‐DM1, Lys‐SMCC‐DM1, had a low level of permeability. Under a coculture condition of HER2‐positive KPL‐4 cells and negative MDA‐MB‐468 cells in vitro, DS‐8201a killed both cells, whereas T‐DM1 and an antibody–drug conjugate with a low permeable payload, anti‐HER2‐DXd (2), did not. In vivo evaluation was carried out using mice inoculated with a mixture of HER2‐positive NCI‐N87 cells and HER2‐negative MDA‐MB‐468‐Luc cells by using an in vivo imaging system. In vivo, DS‐8201a reduced the luciferase signal of the mice, indicating suppression of the MDA‐MB‐468‐Luc population; however, T‐DM1 and anti‐HER2‐DXd (2) did not. Furthermore, it was confirmed that DS‐8201a was not effective against MDA‐MB‐468‐Luc tumors inoculated at the opposite side of the NCI‐N87 tumor, suggesting that the bystander killing effect of DS‐8201a is observed only in cells neighboring HER2‐positive cells, indicating low concern in terms of systemic toxicity. These results indicated that DS‐8201a has a potent bystander effect due to a highly membrane‐permeable payload and is beneficial in treating tumors with HER2 heterogeneity that are unresponsive to T‐DM1.
Collapse
Affiliation(s)
- Yusuke Ogitani
- Biologics & Immuno-Oncology Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Katsunobu Hagihara
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Masataka Oitate
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Hiroyuki Naito
- Oncology Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Toshinori Agatsuma
- Biologics & Immuno-Oncology Laboratories, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
28
|
Ponte JF, Sun X, Yoder NC, Fishkin N, Laleau R, Coccia J, Lanieri L, Bogalhas M, Wang L, Wilhelm S, Widdison W, Pinkas J, Keating TA, Chari R, Erickson HK, Lambert JM. Understanding How the Stability of the Thiol-Maleimide Linkage Impacts the Pharmacokinetics of Lysine-Linked Antibody-Maytansinoid Conjugates. Bioconjug Chem 2016; 27:1588-98. [PMID: 27174129 DOI: 10.1021/acs.bioconjchem.6b00117] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Antibody-drug conjugates (ADCs) have become a widely investigated modality for cancer therapy, in part due to the clinical findings with ado-trastuzumab emtansine (Kadcyla). Ado-trastuzumab emtansine utilizes the Ab-SMCC-DM1 format, in which the thiol-functionalized maytansinoid cytotoxic agent, DM1, is linked to the antibody (Ab) via the maleimide moiety of the heterobifunctional SMCC linker. The pharmacokinetic (PK) data for ado-trastuzumab emtansine point to a faster clearance for the ADC than for total antibody. Cytotoxic agent release in plasma has been reported with nonmaytansinoid, cysteine-linked ADCs via thiol-maleimide exchange, for example, brentuximab vedotin. For Ab-SMCC-DM1 ADCs, however, the main catabolite reported is lysine-SMCC-DM1, the expected product of intracellular antibody proteolysis. To understand these observations better, we conducted a series of studies to examine the stability of the thiol-maleimide linkage, utilizing the EGFR-targeting conjugate, J2898A-SMCC-DM1, and comparing it with a control ADC made with a noncleavable linker that lacked a thiol-maleimide adduct (J2898A-(CH2)3-DM). We employed radiolabeled ADCs to directly measure both the antibody and the ADC components in plasma. The PK properties of the conjugated antibody moiety of the two conjugates, J2898A-SMCC-DM1 and J2898A-(CH2)3-DM (each with an average of 3.0 to 3.4 maytansinoid molecules per antibody), appear to be similar to that of the unconjugated antibody. Clearance values of the intact conjugates were slightly faster than those of the Ab components. Furthermore, J2898A-SMCC-DM1 clears slightly faster than J2898A-(CH2)3-DM, suggesting that there is a fraction of maytansinoid loss from the SMCC-DM1 ADC, possibly through a thiol-maleimide dependent mechanism. Experiments on ex vivo stability confirm that some loss of maytansinoid from Ab-SMCC-DM1 conjugates can occur via thiol elimination, but at a slower rate than the corresponding rate of loss reported for thiol-maleimide links formed at thiols derived by reduction of endogenous cysteine residues in antibodies, consistent with expected differences in thiol-maleimide stability related to thiol pKa. These findings inform the design strategy for future ADCs.
Collapse
Affiliation(s)
- Jose F Ponte
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Xiuxia Sun
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Nicholas C Yoder
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Nathan Fishkin
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Rassol Laleau
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Jennifer Coccia
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Leanne Lanieri
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Megan Bogalhas
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Lintao Wang
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Sharon Wilhelm
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Wayne Widdison
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Jan Pinkas
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Thomas A Keating
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Ravi Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - Hans K Erickson
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| | - John M Lambert
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451-1477, United States
| |
Collapse
|
29
|
Walles M, Rudolph B, Wolf T, Bourgailh J, Suetterlin M, Moenius T, Peraus G, Heudi O, Elbast W, Lanshoeft C, Bilic S. New Insights in Tissue Distribution, Metabolism, and Excretion of [3H]-Labeled Antibody Maytansinoid Conjugates in Female Tumor-Bearing Nude Rats. Drug Metab Dispos 2016; 44:897-910. [DOI: 10.1124/dmd.115.069021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/26/2016] [Indexed: 12/15/2022] Open
|
30
|
Singh R, Setiady YY, Ponte J, Kovtun YV, Lai KC, Hong EE, Fishkin N, Dong L, Jones GE, Coccia JA, Lanieri L, Veale K, Costoplus JA, Skaletskaya A, Gabriel R, Salomon P, Wu R, Qiu Q, Erickson HK, Lambert JM, Chari RVJ, Widdison WC. A New Triglycyl Peptide Linker for Antibody-Drug Conjugates (ADCs) with Improved Targeted Killing of Cancer Cells. Mol Cancer Ther 2016; 15:1311-20. [PMID: 27197308 DOI: 10.1158/1535-7163.mct-16-0021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/19/2016] [Indexed: 11/16/2022]
Abstract
A triglycyl peptide linker (CX) was designed for use in antibody -: drug conjugates (ADC), aiming to provide efficient release and lysosomal efflux of cytotoxic catabolites within targeted cancer cells. ADCs comprising anti-epithelial cell adhesion molecule (anti-EpCAM) and anti-EGFR antibodies with maytansinoid payloads were prepared using CX or a noncleavable SMCC linker (CX and SMCC ADCs). The in vitro cytotoxic activities of CX and SMCC ADCs were similar for several cancer cell lines; however, the CX ADC was more active (5-100-fold lower IC50) than the SMCC ADC in other cell lines, including a multidrug-resistant line. Both CX and SMCC ADCs showed comparable MTDs and pharmacokinetics in CD-1 mice. In Calu-3 tumor xenografts, antitumor efficacy was observed with the anti-EpCAM CX ADC at a 5-fold lower dose than the corresponding SMCC ADC in vivo Similarly, the anti-EGFR CX ADC showed improved antitumor activity over the respective SMCC conjugate in HSC-2 and H1975 tumor models; however, both exhibited similar activity against FaDu xenografts. Mechanistically, in contrast with the charged lysine-linked catabolite of SMCC ADC, a significant fraction of the carboxylic acid catabolite of CX ADC could be uncharged in the acidic lysosomes, and thus diffuse out readily into the cytosol. Upon release from tumor cells, CX catabolites are charged at extracellular pH and do not penetrate and kill neighboring cells, similar to the SMCC catabolite. Overall, these data suggest that CX represents a promising linker option for the development of ADCs with improved therapeutic properties. Mol Cancer Ther; 15(6); 1311-20. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ling Dong
- ImmunoGen, Inc., Waltham, Massachusetts
| | | | | | | | | | | | | | | | | | - Rui Wu
- ImmunoGen, Inc., Waltham, Massachusetts
| | | | | | | | | | | |
Collapse
|
31
|
Force J, Saxena R, Schneider BP, Storniolo AM, Sledge GW, Chalasani N, Vuppalanchi R. Nodular Regenerative Hyperplasia After Treatment With Trastuzumab Emtansine. J Clin Oncol 2016; 34:e9-e12. [DOI: 10.1200/jco.2013.49.8543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Jeremy Force
- Indiana University School of Medicine, Indiana University Health, Indianapolis, IN
| | - Romil Saxena
- Indiana University School of Medicine, Indiana University Health, Indianapolis, IN
| | - Bryan P. Schneider
- Indiana University School of Medicine, Indiana University Health, Indianapolis, IN
| | - Anna M. Storniolo
- Indiana University School of Medicine, Indiana University Health, Indianapolis, IN
| | - George W. Sledge
- Indiana University School of Medicine, Indiana University Health, Indianapolis, IN
| | - Naga Chalasani
- Indiana University School of Medicine, Indiana University Health, Indianapolis, IN
| | - Raj Vuppalanchi
- Indiana University School of Medicine, Indiana University Health, Indianapolis, IN
| |
Collapse
|
32
|
Hamblett KJ, Jacob AP, Gurgel JL, Tometsko ME, Rock BM, Patel SK, Milburn RR, Siu S, Ragan SP, Rock DA, Borths CJ, O'Neill JW, Chang WS, Weidner MF, Bio MM, Quon KC, Fanslow WC. SLC46A3 Is Required to Transport Catabolites of Noncleavable Antibody Maytansine Conjugates from the Lysosome to the Cytoplasm. Cancer Res 2015; 75:5329-40. [PMID: 26631267 DOI: 10.1158/0008-5472.can-15-1610] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/17/2015] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) target cytotoxic drugs to antigen-positive cells for treating cancer. After internalization, ADCs with noncleavable linkers are catabolized to amino acid-linker-warheads within the lysosome, which then enter the cytoplasm by an unknown mechanism. We hypothesized that a lysosomal transporter was responsible for delivering noncleavable ADC catabolites into the cytoplasm. To identify candidate transporters, we performed a phenotypic shRNA screen with an anti-CD70 maytansine-based ADC. This screen revealed the lysosomal membrane protein SLC46A3, the genetic attenuation of which inhibited the potency of multiple noncleavable antibody-maytansine ADCs, including ado-trastuzumab emtansine. In contrast, the potencies of noncleavable ADCs carrying the structurally distinct monomethyl auristatin F were unaffected by SLC46A3 attenuation. Structure-activity experiments suggested that maytansine is a substrate for SLC46A3. Notably, SLC46A3 silencing led to relative increases in catabolite concentrations in the lysosome. Taken together, our results establish SLC46A3 as a direct transporter of maytansine-based catabolites from the lysosome to the cytoplasm, prompting further investigation of SLC46A3 as a predictive response marker in breast cancer specimens.
Collapse
Affiliation(s)
| | - Allison P Jacob
- Amgen Inc., Therapeutic Innovation Unit, Seattle, Washington
| | - Jesse L Gurgel
- Amgen Inc., Therapeutic Innovation Unit, Seattle, Washington
| | - Mark E Tometsko
- Amgen Inc., Therapeutic Innovation Unit, Seattle, Washington
| | - Brooke M Rock
- Amgen Inc., Pharmacokinetics and Drug Metabolism, Seattle, Washington
| | - Sonal K Patel
- Amgen Inc., Pharmacokinetics and Drug Metabolism, Seattle, Washington
| | - Robert R Milburn
- Amgen Inc., Small Molecule Purification and Process Development, Thousand Oaks, California
| | - Sophia Siu
- Amgen Inc., Therapeutic Discovery, Seattle, Washington
| | | | - Dan A Rock
- Amgen Inc., Pharmacokinetics and Drug Metabolism, Seattle, Washington
| | - Christopher J Borths
- Amgen Inc., Small Molecule Purification and Process Development, Thousand Oaks, California
| | | | - Wesley S Chang
- Amgen Inc., Clinical Immunology, South San Francisco, California
| | | | - Matthew M Bio
- Amgen Inc., Small Molecule Purification and Process Development, Thousand Oaks, California
| | - Kim C Quon
- Amgen Inc., Therapeutic Innovation Unit, Seattle, Washington
| | | |
Collapse
|
33
|
Widdison WC, Ponte JF, Coccia JA, Lanieri L, Setiady Y, Dong L, Skaletskaya A, Hong EE, Wu R, Qiu Q, Singh R, Salomon P, Fishkin N, Harris L, Maloney EK, Kovtun Y, Veale K, Wilhelm SD, Audette CA, Costoplus JA, Chari RVJ. Development of Anilino-Maytansinoid ADCs that Efficiently Release Cytotoxic Metabolites in Cancer Cells and Induce High Levels of Bystander Killing. Bioconjug Chem 2015; 26:2261-78. [DOI: 10.1021/acs.bioconjchem.5b00430] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Jose F. Ponte
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | | | - Leanne Lanieri
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Yulius Setiady
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Ling Dong
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | | | - E. Erica Hong
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Rui Wu
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Qifeng Qiu
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Rajeeva Singh
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Paulin Salomon
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Nathan Fishkin
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Luke Harris
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | | | - Yelena Kovtun
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | - Karen Veale
- ImmunoGen Inc., Waltham, Massachusetts 02451, United States
| | | | | | | | | |
Collapse
|
34
|
Abstract
The selective delivery of potent pharmacologically active compounds to target tissue or cells by antibody–drug conjugates makes this immuno-conjugate a promising modality for the treatment of cancers. A thorough understanding of the structural integrity of the linker, the payload and the conjugation site during biological exposure is critical throughout the process of novel linker-payload design and optimization of PK profile. This understanding is a key aspect of the effort to maximize efficacy while minimizing toxicity in preclinical testing and to ensure the translation to the clinical setting. The complexity of this bioconjugate modality is a source of significant challenge for analytical interrogation and analysis in vivo. Therefore, we report herein a survey of various types of biotransformation events that have been elucidated in recent years.
Collapse
|
35
|
Kamath AV, Iyer S. Challenges and advances in the assessment of the disposition of antibody-drug conjugates. Biopharm Drug Dispos 2015; 37:66-74. [PMID: 25904406 PMCID: PMC5032988 DOI: 10.1002/bdd.1957] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/17/2015] [Accepted: 04/01/2015] [Indexed: 11/26/2022]
Abstract
Antibody‐drug conjugates (ADCs) are a rapidly growing therapeutic platform for the treatment of cancer. ADCs consist of a cytotoxic small molecule drug linked to an antibody to provide targeted delivery of the cytotoxic agent to the tumor. Understanding the pharmacokinetics (PK) and pharmacodynamics (PD) of ADCs is crucial in their design to optimize dose and regimen, to maximize efficacy and to minimize toxicity in patients. Significant progress has been made in recent years in this area, however, many fundamental questions still remain. This review discusses factors to consider while assessing the disposition of ADCs, and the unique challenges associated with these therapeutics. Current tools that are available and strategies to enable appropriate assessment are also discussed. © 2015 Genentech Inc. Biopharmaceutics & Drug Disposition Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- Amrita V Kamath
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc, South San Francisco, CA, USA
| | - Suhasini Iyer
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
36
|
Hong EE, Erickson H, Lutz RJ, Whiteman KR, Jones G, Kovtun Y, Blanc V, Lambert JM. Design of Coltuximab Ravtansine, a CD19-Targeting Antibody-Drug Conjugate (ADC) for the Treatment of B-Cell Malignancies: Structure-Activity Relationships and Preclinical Evaluation. Mol Pharm 2015; 12:1703-16. [PMID: 25856201 DOI: 10.1021/acs.molpharmaceut.5b00175] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Coltuximab ravtansine (SAR3419) is an antibody-drug conjugate (ADC) targeting CD19 created by conjugating a derivative of the potent microtubule-acting cytotoxic agent, maytansine, to a version of the anti-CD19 antibody, anti-B4, that was humanized as an IgG1 by variable domain resurfacing. Four different linker-maytansinoid constructs were synthesized (average ∼3.5 maytansinoids/antibody for each) to evaluate the impact of linker-payload design on the activity of the maytansinoid-ADCs targeting CD19. The ADC composed of DM4 (N(2')-deacetyl-N(2')-[4-mercapto-4-methyl-1-oxopentyl]maytansine) conjugated to antibody via the N-succinimidyl-4-(2-pyridyldithio)butyrate (SPDB) linker was selected for development as SAR3419. A molar ratio for DM4/antibody of between 3 and 5 was selected for the final design of SAR3419. Evaluation of SAR3419 in Ramos tumor xenograft models showed that the minimal effective single dose was about 50 μg/kg conjugated DM4 (∼2.5 mg/kg conjugated antibody), while twice this dose gave complete regressions in 100% of the mice. SAR3419 arrests cells in the G2/M phase of the cell cycle, ultimately leading to apoptosis after about 24 h. The results of in vitro and in vivo studies with SAR3419 made with DM4 that was [(3)H]-labeled at the C20 methoxy group of the maytansinoid suggest a mechanism of internalization and intracellular trafficking of SAR3419, ultimately to lysosomes, in which the antibody is fully degraded, releasing lysine-N(ε)-SPDB-DM4 as the initial metabolite. Subsequent intracellular reduction of the disulfide bond between linker and DM4 generates the free thiol species, which is then converted to S-methyl DM4 by cellular methyl transferase activity. We provide evidence to suggest that generation of S-methyl DM4 in tumor cells may contribute to in vivo tumor eradication via bystander killing of neighboring tumor cells. Furthermore, we show that S-methyl DM4 is converted to the sulfoxide and sulfone derivatives in the liver, suggesting that hepatic catabolism of the payload to less cytotoxic maytansinoid species contributes to the overall therapeutic window of SAR3419. This compound is currently in phase II clinical evaluation for the treatment of diffuse large B cell lymphoma.
Collapse
Affiliation(s)
- E Erica Hong
- †ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Hans Erickson
- †ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Robert J Lutz
- †ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Kathleen R Whiteman
- †ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Gregory Jones
- †ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yelena Kovtun
- †ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Veronique Blanc
- §Sanofi, 13 quai Jules Guesde, Vitry sur Seine, 94403 France
| | - John M Lambert
- †ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
37
|
Widdison W, Wilhelm S, Veale K, Costoplus J, Jones G, Audette C, Leece B, Bartle L, Kovtun Y, Chari R. Metabolites of antibody-maytansinoid conjugates: characteristics and in vitro potencies. Mol Pharm 2015; 12:1762-73. [PMID: 25826705 DOI: 10.1021/mp5007757] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Several antibody-maytansinoid conjugates (AMCs) are in clinical trials for the treatment of various cancers. Each of these conjugates can be metabolized by tumor cells to give cytotoxic maytansinoid metabolites that can kill targeted cells. In preclinical studies in mice, the cytotoxic metabolites initially formed in vivo are further processed in the mouse liver to give several oxidized metabolic species. In this work, the primary AMC metabolites were synthesized and incubated with human liver microsomes (HLMs) to determine if human liver would likely give the same metabolites as those formed in mouse liver. The results of these HLM metabolism studies as well as the subsequent syntheses of the resulting HLM oxidation products are presented. Syntheses of the minor impurities formed during the conjugation of AMCs were also conducted to determine their cytotoxicities and to establish how these impurities would be metabolized by HLM.
Collapse
Affiliation(s)
- Wayne Widdison
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Sharon Wilhelm
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Karen Veale
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Juliet Costoplus
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Gregory Jones
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Charlene Audette
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Barbara Leece
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Laura Bartle
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yelena Kovtun
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ravi Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
38
|
Salomon PL, Singh R. Sensitive ELISA Method for the Measurement of Catabolites of Antibody–Drug Conjugates (ADCs) in Target Cancer Cells. Mol Pharm 2015; 12:1752-61. [DOI: 10.1021/acs.molpharmaceut.5b00028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Paulin L. Salomon
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Rajeeva Singh
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
39
|
Fishkin N. Maytansinoid-BODIPY Conjugates: Application to Microscale Determination of Drug Extinction Coefficients and for Quantification of Maytansinoid Analytes. Mol Pharm 2015; 12:1745-51. [PMID: 25738231 DOI: 10.1021/mp500843r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Determining drug to antibody ratios (DAR) for antibody-drug conjugates (ADCs) in early research and development can be hampered by difficulties in accurate weighing of the effector payload and subsequent determination of its extinction coefficient. Two maytansinoids, DM1 and DM4, potent antimitotic agents used in clinical ADCs, were derivatized with the compact fluorophore BODIPY FL using two different linker designs. We identified DM1-mal-BODIPY as a conjugate with little through-space interaction between the maytansinoid and BODIPY chromophores. The 1:1 stoichiometry between the maytansinoid and BODIPY makes the molar concentration of both components equal and the extinction coefficient of the maytansinoid in proportion with the known BODIPY chromophore according to Beer's Law. By only derivatizing 50 μg of unpurified DM1 and analyzing about 25 μg of DM1-mal-BODIPY by UV-vis, we determined εDM1 252 nm and εDM1 280 nm as 26 355 ± 360 and 5230 ± 160 cm(-1) M(-1), respectively. These values are nearly identical to those accepted for DM1 based on weighing >100 mg of pure sample. Surprisingly, some of the maytansinoid-BODIPY conjugates that were synthesized were partially or completely fluorescence-quenched. The green fluorescence of quenched DM4-acetamide-BODIPY could be fully restored in the presence of an antibody designed to tightly bind maytansine. We exploited this observation to develop a simple "mix and read" fluorogenic immunoassay for detection of nanogram quantities of maytansinoids.
Collapse
Affiliation(s)
- Nathan Fishkin
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
40
|
Lai KC, Deckert J, Setiady YY, Shah P, Wang L, Chari R, Lambert JM. Evaluation of Targets for Maytansinoid ADC Therapy Using a Novel Radiochemical Assay. Pharm Res 2015; 32:3593-603. [PMID: 25630819 DOI: 10.1007/s11095-015-1633-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/16/2015] [Indexed: 10/24/2022]
Abstract
PURPOSE Many antibody-drug conjugates (ADCs) become active only after antigen-mediated internalization and release of the cytotoxic agent via antibody degradation. Quantifying these processes can provide critical information on the suitability of a particular receptor target or antibody for ADC therapy by providing insight into the amount of cytotoxic agent released. We describe a simple and inexpensive radiolabel assay to monitor this process in cultured cancer cells. METHODS Monoclonal antibodies were trace-labeled at their lysine residues by treatment with the N-hydroxysuccinimide ester of [(3)H]propionic acid. Human cancer cell cultures were treated with the labeled antibody at concentrations sufficient to saturate the targeted antigen. After washing to remove unbound antibody, cells were incubated and analyzed for antigen expression, conjugate degradation and catabolite formation. Results were compared with data obtained from similar assays run with radiolabeled antibody-[(3)H]maytansinoid conjugates ([(3)H]AMCs). To exemplify the method, studies were conducted with a panel of [(3)H]propionamide-antibodies to evaluate processing efficiency in EGFR-expressing SCCHN cell lines, and in NHL cell lines expressing the B-cell targets CD19, CD20, CD22 and CD37. RESULTS Use of the [(3)H]propionamide-antibody assay yielded cell-mediated processing results similar to those obtained with corresponding maytansinoid ADCs. Further exploration allowed comparison of expression levels, antigen-dependent degradation, and catabolite formation across a panel of EGFR-expressing SCCHN cell lines, and for multiple targets in various B-cell cancer indications. CONCLUSIONS The [(3)H]propionamide-antibody assay described here is a sensitive, facile method which enables rapid and robust assessment of relative antibody processing amounts for target, antibody, and cell line evaluation.
Collapse
Affiliation(s)
| | | | | | - Prerak Shah
- ImmunoGen, Inc., Waltham, Massachusetts, USA
| | - Lintao Wang
- ImmunoGen, Inc., Waltham, Massachusetts, USA
| | - Ravi Chari
- ImmunoGen, Inc., Waltham, Massachusetts, USA
| | | |
Collapse
|
41
|
Kamath AV, Iyer S. Preclinical Pharmacokinetic Considerations for the Development of Antibody Drug Conjugates. Pharm Res 2014; 32:3470-9. [PMID: 25446773 PMCID: PMC4596897 DOI: 10.1007/s11095-014-1584-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 11/20/2014] [Indexed: 11/29/2022]
Abstract
Antibody drug conjugates (ADCs) are an emerging new class of targeted therapeutics for cancer that use antibodies to deliver cytotoxic drugs to cancer cells. There are two FDA approved ADCs on the market and over 30 ADCs in the clinical pipeline against a number of different cancer types. The structure of an ADC is very complex with multiple components and considerable efforts are ongoing to determine the attributes necessary for clinical success. Understanding the pharmacokinetics of an ADC and how it impacts efficacy and toxicity is a critical part of optimizing ADC design and delivery i.e., dose and schedule. This review discusses the pharmacokinetic considerations for an ADC and tools and strategies that can be used to evaluate molecules at the preclinical stage.
Collapse
Affiliation(s)
- Amrita V Kamath
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc, 1 DNA Way (Mailstop 463A), South San Francisco, CA, 94080, USA.
| | - Suhasini Iyer
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc, 1 DNA Way (Mailstop 463A), South San Francisco, CA, 94080, USA
| |
Collapse
|
42
|
Albers AE, Garofalo AW, Drake PM, Kudirka R, de Hart GW, Barfield RM, Baker J, Banas S, Rabuka D. Exploring the effects of linker composition on site-specifically modified antibody-drug conjugates. Eur J Med Chem 2014; 88:3-9. [PMID: 25176286 DOI: 10.1016/j.ejmech.2014.08.062] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 11/24/2022]
Abstract
In the context of antibody-drug conjugates (ADCs), noncleavable linkers provide a means to deliver cytotoxic small molecules to cell targets while reducing systemic toxicity caused by nontargeted release of the free drug. Additionally, noncleavable linkers afford an opportunity to change the chemical properties of the small molecule to improve potency or diminish affinity for multidrug transporters, thereby improving efficacy. We employed the aldehyde tag coupled with the hydrazino-iso-Pictet-Spengler (HIPS) ligation to generate a panel of site-specifically conjugated ADCs that varied only in the noncleavable linker portion. The ADC panel comprised antibodies carrying a maytansine payload ligated through one of five different linkers. Both the linker-maytansine constructs alone and the resulting ADC panel were characterized in a variety of in vitro and in vivo assays measuring biophysical and functional properties. We observed that slight differences in linker design affected these parameters in disparate ways, and noted that efficacy could be improved by selecting for particular attributes. These studies serve as a starting point for the exploration of more potent noncleavable linker systems.
Collapse
Affiliation(s)
- Aaron E Albers
- Redwood Bioscience, 5703 Hollis Street, Emeryville, CA 94608, USA
| | | | - Penelope M Drake
- Redwood Bioscience, 5703 Hollis Street, Emeryville, CA 94608, USA
| | - Romas Kudirka
- Redwood Bioscience, 5703 Hollis Street, Emeryville, CA 94608, USA
| | | | - Robyn M Barfield
- Redwood Bioscience, 5703 Hollis Street, Emeryville, CA 94608, USA
| | - Jeanne Baker
- Redwood Bioscience, 5703 Hollis Street, Emeryville, CA 94608, USA
| | - Stefanie Banas
- Redwood Bioscience, 5703 Hollis Street, Emeryville, CA 94608, USA
| | - David Rabuka
- Redwood Bioscience, 5703 Hollis Street, Emeryville, CA 94608, USA.
| |
Collapse
|
43
|
Han TH, Zhao B. Absorption, distribution, metabolism, and excretion considerations for the development of antibody-drug conjugates. Drug Metab Dispos 2014; 42:1914-20. [PMID: 25048520 DOI: 10.1124/dmd.114.058586] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a class of therapeutics that are designed to deliver potent small-molecule drugs selectively to cells that express a specific target antigen while limiting systemic exposure to the drug. This is accomplished by conjugating a potent drug onto an antibody-based therapeutic with a linker that is exquisitely stable in plasma. The development of an effective ADC requires optimizing a number of design elements and an extensive understanding of absorption, distribution, metabolism/catabolism, and elimination (ADME) processes for the ADC construct. Furthermore, as ADCs are a combination of an antibody and small-molecule drug, understanding key aspects of the ADME of each individual component is needed. This review aims to provide considerations for the development of ADCs from an ADME point of view.
Collapse
Affiliation(s)
- Tae H Han
- Stem CentRx, Inc. (T.H.H.), South San Francisco, California; Seattle Genetics, Inc. (B.Z.), Bothell, Washington
| | - Baiteng Zhao
- Stem CentRx, Inc. (T.H.H.), South San Francisco, California; Seattle Genetics, Inc. (B.Z.), Bothell, Washington
| |
Collapse
|
44
|
Lambert JM, Chari RVJ. Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J Med Chem 2014; 57:6949-64. [PMID: 24967516 DOI: 10.1021/jm500766w] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ado-trastuzumab emtansine (T-DM1) is an antibody-drug conjugate that combines the antitumor properties of the humanized anti-human epidermal growth factor receptor 2 (HER2) antibody, trastuzumab, with the maytansinoid, DM1, a potent microtubule-disrupting agent, joined by a stable linker. Upon binding to HER2, the conjugate is internalized via receptor-mediated endocytosis, and an active derivative of DM1 is subsequently released by proteolytic degradation of the antibody moiety within the lysosome. Initial clinical evaluation led to a phase III trial in advanced HER2-positive breast cancer patients who had relapsed after prior treatment with trastuzumab and a taxane, which showed that T-DM1 significantly prolonged progression-free and overall survival with less toxicity than lapatinib plus capecitabine. In 2013, T-DM1 received FDA approval for the treatment of patients with HER2-positive metastatic breast cancer who had previously received trastuzumab and a taxane, separately or in combination, the first ADC to receive full approval based on a randomized study.
Collapse
Affiliation(s)
- John M Lambert
- ImmunoGen, Inc. , 830 Winter Street, Waltham, Massachusetts 02451, United States
| | | |
Collapse
|
45
|
Sauerborn M, van Dongen W. Practical Considerations for the Pharmacokinetic and Immunogenic Assessment of Antibody–Drug Conjugates. BioDrugs 2014; 28:383-91. [DOI: 10.1007/s40259-014-0096-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
46
|
Krall N, Pretto F, Decurtins W, Bernardes GJL, Supuran CT, Neri D. A Small-Molecule Drug Conjugate for the Treatment of Carbonic Anhydrase IX Expressing Tumors. Angew Chem Int Ed Engl 2014; 53:4231-5. [DOI: 10.1002/anie.201310709] [Citation(s) in RCA: 211] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Indexed: 11/07/2022]
|
47
|
Ein niedermolekulares Ligand-Wirkstoff-Konjugat zur Behandlung von Carboanhydrase IX exprimierenden Tumoren. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201310709] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
48
|
Chari RVJ, Miller ML, Widdison WC. Antibody-drug conjugates: an emerging concept in cancer therapy. Angew Chem Int Ed Engl 2014; 53:3796-827. [PMID: 24677743 DOI: 10.1002/anie.201307628] [Citation(s) in RCA: 711] [Impact Index Per Article: 71.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Indexed: 01/17/2023]
Abstract
Traditional cancer chemotherapy is often accompanied by systemic toxicity to the patient. Monoclonal antibodies against antigens on cancer cells offer an alternative tumor-selective treatment approach. However, most monoclonal antibodies are not sufficiently potent to be therapeutically active on their own. Antibody-drug conjugates (ADCs) use antibodies to deliver a potent cytotoxic compound selectively to tumor cells, thus improving the therapeutic index of chemotherapeutic agents. The recent approval of two ADCs, brentuximab vedotin and ado-trastuzumab emtansine, for cancer treatment has spurred tremendous research interest in this field. This Review touches upon the early efforts in the field, and describes how the lessons learned from the first-generation ADCs have led to improvements in every aspect of this technology, i.e., the antibody, the cytotoxic compound, and the linker connecting them, leading to the current successes. The design of ADCs currently in clinical development, and results from mechanistic studies and preclinical and clinical evaluation are discussed. Emerging technologies that seek to further advance this exciting area of research are also discussed.
Collapse
Affiliation(s)
- Ravi V J Chari
- ImmunoGen, Inc. 830 Winter St, Waltham, MA 02451 (USA) http://www.immunogen.com.
| | | | | |
Collapse
|
49
|
Chari RVJ, Miller ML, Widdison WC. Antikörper-Wirkstoff-Konjugate: ein neues Konzept in der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201307628] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
50
|
Perrino E, Steiner M, Krall N, Bernardes GJ, Pretto F, Casi G, Neri D. Curative Properties of Noninternalizing Antibody–Drug Conjugates Based on Maytansinoids. Cancer Res 2014; 74:2569-78. [DOI: 10.1158/0008-5472.can-13-2990] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|