1
|
Tamemoto Y, Nakamura Y, Kurino T, Tang R, Arai T, Yasuda S, Kume R, Suzuki H, Uehara T, Akita H, Hisaka A, Hatakeyama H. Antibody-dependent cellular cytotoxicity of anti-programmed death-ligand 1 antibodies for T cells attenuate their antitumor efficacy in a murine tumor model. Int J Pharm 2025; 679:125755. [PMID: 40412452 DOI: 10.1016/j.ijpharm.2025.125755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 05/15/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
We evaluated the significant determinants of the therapeutic efficacy of anti-programmed death-ligand 1 (aPD-L1) monoclonal antibodies (aPD-L1 mAbs) by comparing the differences in antibody-dependent cellular cytotoxicity (ADCC) activity of aPD-L1 mAbs of different isotypes (clones 10F.9G2 and MIH6). In MM48 tumor-bearing mice, MIH6 without ADCC activity inhibited tumor growth by more than 90 %, whereas 10F.9G2 with ADCC activity showed slight antitumor effect. The dissociation constant of PD-L1 on MM48 cell surface was approximately 100-fold lower for MIH6 than for 10F.9G2. However, antibody uptake by cells was comparable for both aPD-L1 mAbs. The half-life and amount of intact form of aPD-L1 mAbs in tumors were 2.6- and 1.6-fold higher in MIH6 cells than in 10F.9G2 cells, respectively. Therefore, differences in the pharmacokinetics of PD-L1 mAbs along with different ADCC activities are insufficient to explain their different antitumor effects. Only 10F.9G2 reduced the number of CD8+ T cells in MM48 tumor-bearing mice. These results suggest that the difference in efficacy of aPD-L1 mAbs could be because aPD-L1 mAbs recognize PD-L1 on T cells, and the number of CD8+ T cells exerting antitumor effect is depleted by 10F.9G2 through ADCC activity. In conclusion, the effect of differences in the pharmacokinetics of aPD-L1 mAbs on the antitumor effect in MM48 tumor-bearing mice was considered negligible. Additionally, the difference in ADCC activity of aPD-L1 mAbs caused a decrease in CD8+ T cells, and this off-target effect was considered a factor affecting their antitumor effects.
Collapse
Affiliation(s)
- Yuta Tamemoto
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan; Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Yoshito Nakamura
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan; Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Taiki Kurino
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Ruiheng Tang
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Takahiro Arai
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Shogo Yasuda
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Riho Kume
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Hiroyuki Suzuki
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Tomoya Uehara
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai City, Miyagi 980-8578, Japan
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan
| | - Hiroto Hatakeyama
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan; Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-City, Chiba 260-8675, Japan.
| |
Collapse
|
2
|
Datta-Mannan A, Choi H, Jin Z, Liu L, Lu J, Stokell DJ, Murphy AT, Dunn KW, Martinez MM, Feng Y. Reducing target binding affinity improves the therapeutic index of anti-MET antibody-drug conjugate in tumor bearing animals. PLoS One 2024; 19:e0293703. [PMID: 38630694 PMCID: PMC11023234 DOI: 10.1371/journal.pone.0293703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/17/2023] [Indexed: 04/19/2024] Open
Abstract
Many oncology antibody-drug conjugates (ADCs) have failed to demonstrate efficacy in clinic because of dose-limiting toxicity caused by uptake into healthy tissues. We developed an approach that harnesses ADC affinity to broaden the therapeutic index (TI) using two anti-mesenchymal-epithelial transition factor (MET) monoclonal antibodies (mAbs) with high affinity (HAV) or low affinity (LAV) conjugated to monomethyl auristatin E (MMAE). The estimated TI for LAV-ADC was at least 3 times greater than the HAV-ADC. The LAV- and HAV-ADCs showed similar levels of anti-tumor activity in the xenograft model, while the 111In-DTPA studies showed similar amounts of the ADCs in HT29 tumors. Although the LAV-ADC has ~2-fold slower blood clearance than the HAV-ADC, higher liver toxicity was observed with HAV-ADC. While the SPECT/CT 111In- and 124I- DTPA findings showed HAV-ADC has higher accumulation and rapid clearance in normal tissues, intravital microscopy (IVM) studies confirmed HAV mAb accumulates within hepatic sinusoidal endothelial cells while the LAV mAb does not. These results demonstrated that lowering the MET binding affinity provides a larger TI for MET-ADC. Decreasing the affinity of the ADC reduces the target mediated drug disposition (TMDD) to MET expressed in normal tissues while maintaining uptake/delivery to the tumor. This approach can be applied to multiple ADCs to improve the clinical outcomes.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Exploratory Medicine and Pharmacology, Lilly Corporate Center, Indianapolis, IN, United States of America
| | - Hiuwan Choi
- Bioproduct Research & Development, Lilly Technology Center North, Indianapolis, IN, United States of America
| | - Zhaoyan Jin
- Drug Disposition/Commercialization, Lilly Corporate Center, Indianapolis, IN, United States of America
| | - Ling Liu
- Biotechnology Discovery Research, Lilly Technology Center North, Indianapolis, IN, United States of America
| | - Jirong Lu
- Biotechnology Discovery Research, Lilly Technology Center North, Indianapolis, IN, United States of America
| | - David J. Stokell
- Biotechnology Discovery Research, Lilly Technology Center North, Indianapolis, IN, United States of America
| | - Anthony T. Murphy
- Drug Disposition/Commercialization, Lilly Corporate Center, Indianapolis, IN, United States of America
| | - Kenneth W. Dunn
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Michelle M. Martinez
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Yiqing Feng
- Biotechnology Discovery Research, Lilly Technology Center North, Indianapolis, IN, United States of America
| |
Collapse
|
3
|
Sandker GGW, Middelburg J, Wilbrink E, Molkenboer-Kuenen J, Aarntzen E, van Hall T, Heskamp S. Longitudinal evaluation of the biodistribution and cellular internalization of the bispecific CD3xTRP1 antibody in syngeneic mouse tumor models. J Immunother Cancer 2023; 11:e007596. [PMID: 37899133 PMCID: PMC10619024 DOI: 10.1136/jitc-2023-007596] [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] [Accepted: 10/04/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND CD3 bispecific antibodies (CD3-bsAbs) require binding of both a tumor-associated surface antigen and CD3 for their immunotherapeutic effect. Their efficacy is, therefore, influenced by the tumor uptake and the extracellular dose. To optimize their currently limited efficacy in solid tumors, increased understanding of their pharmacokinetics and in vivo internalization is needed. METHODS Here, were studied the pharmacokinetics and in vivo internalization of CD3xTRP1, a fully murine Fc-inert bsAb, in endogenous TRP1-expressing immunocompetent male C57BL/6J mice bearing TRP1-positive and negative tumors over time. Matching bsAbs lacking TRP1-binding or CD3-binding capacity served as controls. BsAbs were radiolabeled with 111In to investigate their pharmacokinetics, target binding, and biodistribution through SPECT/CT imaging and ex vivo biodistribution analyses. Co-injection of 111In- and 125I-labeled bsAb was performed to investigate the in vivo internalization by comparing tissue concentrations of cellular residing 111In versus effluxing 125I. Antitumor therapy effects were evaluated by monitoring tumor growth and immunohistochemistry. RESULTS SPECT/CT and biodistribution analyses showed that CD3xTRP1 specifically targeted TRP1-positive tumors and CD3-rich lymphoid organ and uptake peaked 24 hours pi (KPC3-TRP1: 37.7%ID/g±5.3%ID/g, spleen: 29.0%ID/g±3.9%ID/g). Studies with control bsAbs demonstrated that uptake of CD3xTRP1 in TRP1-positive tumors and CD3-rich tissues was primarily receptor-mediated. Together with CD3xTRP1 in the circulation being mainly unattached, this indicates that CD3+ T cells are generally not traffickers of CD3-bsAbs to the tumor. Additionally, target-mediated clearance by TRP1-expressing melanocytes was not observed. We further demonstrated rapid internalization of CD3xTRP1 in KPC3-TRP1 tumors (24 hours pi: 54.9%±2.3% internalized) and CD3-rich tissues (spleen, 24 hours pi: 79.7%±0.9% internalized). Therapeutic effects by CD3xTRP1 were observed for TRP1-positive tumors and consisted of high tumor influx of CD8+ T cells and neutrophils, which corresponded with increased necrosis and growth delay. CONCLUSIONS We show that CD3xTRP1 efficiently targets TRP1-positive tumors and CD3-rich tissues primarily through receptor-mediated targeting. We further demonstrate rapid receptor-mediated internalization of CD3xTRP1 in TRP1-positive tumors and CD3-rich tissues. Even though this significantly decreases the therapeutical available dose, CD3xTRP1 still induced effective antitumor T-cell responses and inhibited tumor growth. Together, our data on the pharmacokinetics and mechanism of action of CD3xTRP1 pave the way for further optimization of CD3-bsAb therapies.
Collapse
Affiliation(s)
| | - Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Evienne Wilbrink
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Erik Aarntzen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
4
|
Datta-Mannan A, Molitoris BA, Feng Y, Martinez MM, Sandoval RM, Brown RM, Merkel D, Croy JE, Dunn KW. Intravital Microscopy Reveals Unforeseen Biodistribution Within the Liver and Kidney Mechanistically Connected to the Clearance of a Bifunctional Antibody. Drug Metab Dispos 2023; 51:403-412. [PMID: 36460476 PMCID: PMC11022859 DOI: 10.1124/dmd.122.001049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/16/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Bifunctional antibody (BfAb) therapeutics offer the potential for novel functionalities beyond those of the individual monospecific entities. However, combining these entities into a single molecule can have unpredictable effects, including changes in pharmacokinetics that limit the compound's therapeutic profile. A better understanding of how molecular modifications affect in vivo tissue interactions could help inform BfAb design. The present studies were predicated on the observation that a BfAb designed to have minimal off-target interactions cleared from the circulation twice as fast as the monoclonal antibody (mAb) from which it was derived. The present study leverages the spatial and temporal resolution of intravital microscopy (IVM) to identify cellular interactions that may explain the different pharmacokinetics of the two compounds. Disposition studies of mice demonstrated that radiolabeled compounds distributed similarly over the first 24 hours, except that BfAb accumulated approximately two- to -three times more than mAb in the liver. IVM studies of mice demonstrated that both distributed to endosomes of liver endothelia but with different kinetics. Whereas mAb accumulated rapidly within the first hour of administration, BfAb accumulated only modestly during the first hour but continued to accumulate over 24 hours, ultimately reaching levels similar to those of the mAb. Although neither compound was freely filtered by the mouse or rat kidney, BfAb, but not mAb, was found to accumulate over 24 hours in endosomes of proximal tubule cells. These studies demonstrate how IVM can be used as a tool in drug design, revealing unpredicted cellular interactions that are undetectable by conventional analyses. SIGNIFICANCE STATEMENT: Bifunctional antibodies offer novel therapeutic functionalities beyond those of the individual monospecific entities. However, combining these entities into a single molecule can have unpredictable effects, including undesirable changes in pharmacokinetics. Studies of the dynamic distribution of a bifunctional antibody and its parent monoclonal antibody presented here demonstrate how intravital microscopy can expand our understanding of the in vivo disposition of therapeutics, detecting off-target interactions that could not be detected by conventional pharmacokinetics approaches or predicted by conventional physicochemical analyses.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Bruce A Molitoris
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Yiqing Feng
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Michelle M Martinez
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Ruben M Sandoval
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Robin M Brown
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Daniel Merkel
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Johnny E Croy
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Kenneth W Dunn
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| |
Collapse
|
5
|
Chang HP, Li Z, Shah DK. Development of a Physiologically-Based Pharmacokinetic Model for Whole-Body Disposition of MMAE Containing Antibody-Drug Conjugate in Mice. Pharm Res 2022; 39:1-24. [PMID: 35044590 DOI: 10.1007/s11095-021-03162-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE To quantitate and mathematically characterize the whole-body pharmacokinetics (PK) of different ADC analytes following administration of an MMAE-conjugated ADC in tumor-bearing mice. METHODS The PK of different ADC analytes (total antibody, total drug, unconjugated drug) was measured following administration of an MMAE-conjugated ADC in tumor-bearing mice. The PK of ADC analytes was compared with the whole-body PK of the antibody and drug obtained following administration of these molecules alone. An ADC PBPK model was developed by linking antibody PBPK model with small-molecule PBPK model, where the drug was assumed to deconjugate in DAR-dependent manner. RESULTS Comparison of antibody biodistribution coefficient (ABC) values for total antibody suggests that conjugation of drug did not significantly affect the PK of antibody. Comparison of tissue:plasma AUC ratio (T/P) for the conjugated drug and total antibody suggests that in certain tissues (e.g., spleen) ADC may demonstrate higher deconjugation. It was observed that the tissue distribution profile of the drug can be altered following its conjugation to antibody. For example, MMAE distribution to the liver was found to increase while its distribution to the heart was found to decrease upon conjugation to antibody. MMAE exposure in the tumor was found to increase by ~20-fold following administration as conjugate (i.e., ADC). The PBPK model was able to a priori predict the PK of all three ADC analytes in plasma, tissues, and tumor reasonably well. CONCLUSIONS The ADC PBPK model developed here serves as a platform for translational and clinical investigations of MMAE containing ADCs.
Collapse
Affiliation(s)
- Hsuan-Ping Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Zhe Li
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
6
|
Kurino T, Matsuda R, Terui A, Suzuki H, Kokubo T, Uehara T, Arano Y, Hisaka A, Hatakeyama H. Poor outcome with anti-programmed death-ligand 1 (PD-L1) antibody due to poor pharmacokinetic properties in PD-1/PD-L1 blockade-sensitive mouse models. J Immunother Cancer 2021; 8:jitc-2019-000400. [PMID: 32041818 PMCID: PMC7057431 DOI: 10.1136/jitc-2019-000400] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2019] [Indexed: 01/08/2023] Open
Abstract
Background Recently, antiprogrammed cell death protein 1 (aPD-1) and antiprogrammed death-ligand 1 (aPD-L1) monoclonal antibodies (mAbs) have been approved. Even though aPD-1 and aPD-L1 mAbs target the same PD-1/PD-L1 axis, it is still unclear whether both mAbs exert equivalent pharmacological activity in patients who are sensitive to PD-1/PD-L1 blockade therapy, as there is no direct comparison of their pharmacokinetics (PK) and antitumor effects. Therefore, we evaluated the differences between both mAbs in PK and therapeutic effects in PD-1/PD-L1 blockade-sensitive mouse models. Methods Herein, murine breast MM48 and colon MC38 xenografts were used to analyze the pharmacological activity of aPD-1 and aPD-L1 mAbs. The PK of the mAbs in the tumor-bearing mice was investigated at low and high doses using two radioisotopes (Indium-111 and Iodine-125) to evaluate the accumulation and degradation of the mAbs. Results aPD-1 mAb showed antitumor effect in a dose-dependent manner, indicating that the tumor model was sensitive to PD-1/PD-L1 blockade therapy, whereas aPD-L1 mAb failed to suppress tumor growth. The PK study showed that aPD-L1 mAb was accumulated largely in normal organs such as the spleen, liver, and kidney, resulting in low blood concentration and low distributions to tumors at a low dose, even though the tumors expressed PD-L1. Sufficient accumulation of aPD-L1 mAb in tumors was achieved by administration at a high dose owing to the saturation of target-mediated binding in healthy organs. However, degradation of aPD-L1 mAb in tumors was greater than that of aPD-1 mAb, which resulted in poor outcome presumably due to less inhibition of PD-L1 by aPD-L1 mAb than that of PD-1 by aPD-1 mAb. Conclusion According to the PK studies, aPD-1 mAb showed linear PK, whereas aPD-L1 mAb showed non-linear PK between low and high doses. Collectively, the poor PK characteristics of aPD-L1 mAb caused lower antitumor activity than of aPD-1 mAb. These results clearly indicated that aPD-L1 mAb required higher doses than aPD-1 mAb in clinical setting. Thus, targeting of PD-1 would be more advantageous than PD-L1 in terms of PK.
Collapse
Affiliation(s)
- Taiki Kurino
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Reiko Matsuda
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Ayu Terui
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroyuki Suzuki
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Tomomi Kokubo
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Tomoya Uehara
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Yasushi Arano
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Hiroto Hatakeyama
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
7
|
Preclinical safety profile of disitamab vedotin:a novel anti-HER2 antibody conjugated with MMAE. Toxicol Lett 2019; 324:30-37. [PMID: 31877330 DOI: 10.1016/j.toxlet.2019.12.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 11/21/2022]
Abstract
The HER2 pathway plays a pivotal role in cell proliferation and differentiation, while the receptor overexpression caused by amplification of HER2 gene is associated with the growth of several tumors. Previously published clinical trials have demonstrated that antibody-conjugated drugs (ADCs) remarkably improved clinical effects compared with antibodies alone for the same target. In order to provide more effective drugs, we developed Disitamab vedotin based on ADC. The antibody part was a humanized monoclonal antibody targeting HER2, the small molecule toxin was monomethyl auristatin E (MMAE), a synthetic antineoplastic agent. A protease cleavable linker covalently attached MMAE to the antibody. In this study, we characterized the toxicity profile of Disitamab vedotin through single- and repeat-dose toxicity studies in monkeys. The toxicities of small molecules and naked antibody (Disitamab) were also assessed in these studies. Monkeys were well tolerated with Disitamab vedotin at doses of 6 mg/kg, while equivalent MMAEs resulted in severe myelosuppression. This finding proves that ADCs improve the therapeutic effect. In addition, the safety profiles of Disitamab vedotin and MMAE were similar and consistent with the activation mechanism of MMAE. Toxicology finding included bone marrow/hematology toxicity and lymphoid organ toxicity, while no significant toxicity was observed in animals treated with naked antibody. These side effects were found to be consistent with data acquired from clinical phase I/II patients treated with Disitamab vedotin.
Collapse
|
8
|
Boswell CA, Yadav DB, Mundo EE, Yu SF, Lacap JA, Fourie-O'Donohue A, Kozak KR, Ferl GZ, Zhang C, Ho J, Ulufatu S, Khawli LA, Lin K. Biodistribution and efficacy of an anti-TENB2 antibody-drug conjugate in a patient-derived model of prostate cancer. Oncotarget 2019; 10:6234-6244. [PMID: 31692898 PMCID: PMC6817444 DOI: 10.18632/oncotarget.27263] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/01/2019] [Indexed: 01/08/2023] Open
Abstract
TENB2, a transmembrane proteoglycan protein, is a promising target for antibody drug conjugate (ADC) therapy due to overexpression in human prostate tumors and rapid internalization. We previously characterized how predosing with parental anti-TENB2 monoclonal antibody (mAb) at 1 mg/kg in a patient-derived LuCap77 explant model with high (3+) TENB2 expression could (i) block target-mediated intestinal uptake of tracer (& 0.1 mg/kg) levels of radiolabeled anti-TENB2-monomethyl auristatin E ADC while preserving tumor uptake, and (ii) maintain efficacy relative to ADC alone. Here, we systematically revisit this strategy to evaluate the effects of predosing on tumor uptake and efficacy in LuCap96.1, a low TENB2-expressing (1+) patient-derived model that is more responsive to ADC therapy than LuCap77. Importantly, rather than using tracer (& 0.1 mg/kg) levels, radiolabeled ADC tumor uptake was assessed at 1 mg/kg – one of the doses evaluated in the tumor growth inhibition study – in an effort to bridge tissue distribution (PK) with efficacy (PD). Predosing with mAb up to 1 mg/kg had no effect on efficacy. These findings warrant further investigations to determine whether predosing prior to ADC therapy might improve therapeutic index by preventing ADC disposition and possible toxicological liabilities in antigen-expressing healthy tissues.
Collapse
Affiliation(s)
- C Andrew Boswell
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | | | - Eduardo E Mundo
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA.,Present address: Department of Safety Assessment, Nektar Therapeutics, San Francisco, 94158 CA, USA
| | - Shang-Fan Yu
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | - Jennifer Arca Lacap
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | | | - Katherine R Kozak
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | - Gregory Z Ferl
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | - Crystal Zhang
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | - Jason Ho
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | - Sheila Ulufatu
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA
| | - Leslie A Khawli
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA.,Present address: Department of Pathology and Laboratory Medicine, Keck School of Medicine of USC, Los Angeles, 90033 CA, USA
| | - Kedan Lin
- Genentech Research and Early Development, South San Francisco, 94080 CA, USA.,Present address: Clinical Development and US Operation, Innovent Biologics, South San Francisco, 94080 CA, USA
| |
Collapse
|
9
|
Lombana TN, Rajan S, Zorn JA, Mandikian D, Chen EC, Estevez A, Yip V, Bravo DD, Phung W, Farahi F, Viajar S, Lee S, Gill A, Sandoval W, Wang J, Ciferri C, Boswell CA, Matsumoto ML, Spiess C. Production, characterization, and in vivo half-life extension of polymeric IgA molecules in mice. MAbs 2019; 11:1122-1138. [PMID: 31122132 PMCID: PMC6748581 DOI: 10.1080/19420862.2019.1622940] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IgA antibodies have broad potential as a novel therapeutic platform based on their superior receptor-mediated cytotoxic activity, potent neutralization of pathogens, and ability to transcytose across mucosal barriers via polymeric immunoglobulin receptor (pIgR)-mediated transport, compared to traditional IgG-based drugs. However, the transition of IgA into clinical development has been challenged by complex expression and characterization, as well as rapid serum clearance that is thought to be mediated by glycan receptor scavenging of recombinantly produced IgA monomer bearing incompletely sialylated N-linked glycans. Here, we present a comprehensive biochemical, biophysical, and structural characterization of recombinantly produced monomeric, dimeric and polymeric human IgA. We further explore two strategies to overcome the rapid serum clearance of polymeric IgA: removal of all N-linked glycosylation sites creating an aglycosylated polymeric IgA and engineering in FcRn binding with the generation of a polymeric IgG-IgA Fc fusion. While previous reports and the results presented in this study indicate that glycan-mediated clearance plays a major role for monomeric IgA, systemic clearance of polymeric IgA in mice is predominantly controlled by mechanisms other than glycan receptor clearance, such as pIgR-mediated transcytosis. The developed IgA platform now provides the potential to specifically target pIgR expressing tissues, while maintaining low systemic exposure.
Collapse
Affiliation(s)
- T Noelle Lombana
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Sharmila Rajan
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Julie A Zorn
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - Danielle Mandikian
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Eugene C Chen
- d Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Alberto Estevez
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - Victor Yip
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Daniel D Bravo
- e Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco , CA , USA
| | - Wilson Phung
- f Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc. , South San Francisco , CA , USA
| | - Farzam Farahi
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Sharon Viajar
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Sophia Lee
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Avinash Gill
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| | - Wendy Sandoval
- f Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc. , South San Francisco , CA , USA
| | - Jianyong Wang
- e Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco , CA , USA
| | - Claudio Ciferri
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - C Andrew Boswell
- b Department of Preclinical and Translational Pharmacokinetics, Genentech Inc., South San Francisco , CA , USA
| | - Marissa L Matsumoto
- c Department of Structural Biology, Genentech Inc., South San Francisco , CA , USA
| | - Christoph Spiess
- a Department of Antibody Engineering, Genentech Inc., South San Francisco , CA , USA
| |
Collapse
|
10
|
Nejadmoghaddam MR, Minai-Tehrani A, Ghahremanzadeh R, Mahmoudi M, Dinarvand R, Zarnani AH. Antibody-Drug Conjugates: Possibilities and Challenges. Avicenna J Med Biotechnol 2019; 11:3-23. [PMID: 30800238 PMCID: PMC6359697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 12/31/2017] [Indexed: 11/22/2022] Open
Abstract
The design of Antibody Drug Conjugates (ADCs) as efficient targeting agents for tumor cell is still in its infancy for clinical applications. This approach incorporates the antibody specificity and cell killing activity of chemically conjugated cytotoxic agents. Antibody in ADC structure acts as a targeting agent and a nanoscale carrier to deliver a therapeutic dose of cytotoxic cargo into desired tumor cells. Early ADCs encountered major obstacles including, low blood residency time, low penetration capacity to tumor microenvironment, low payload potency, immunogenicity, unusual off-target toxicity, drug resistance, and the lack of stable linkage in blood circulation. Although extensive studies have been conducted to overcome these issues, the ADCs based therapies are still far from having high-efficient clinical outcomes. This review outlines the key characteristics of ADCs including tumor marker, antibody, cytotoxic payload, and linkage strategy with a focus on technical improvement and some future trends in the pipeline.
Collapse
Affiliation(s)
- Mohammad-Reza Nejadmoghaddam
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Arash Minai-Tehrani
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Ramin Ghahremanzadeh
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Morteza Mahmoudi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Immunology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
| |
Collapse
|
11
|
Endo Y, Takeda K, Mohan N, Shen Y, Jiang J, Rotstein D, Wu WJ. Payload of T-DM1 binds to cell surface cytoskeleton-associated protein 5 to mediate cytotoxicity of hepatocytes. Oncotarget 2018; 9:37200-37215. [PMID: 30647854 PMCID: PMC6324681 DOI: 10.18632/oncotarget.26461] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022] Open
Abstract
Off-target toxicity is a major cause of dose-limiting toxicity for antibody-drug conjugates (ADCs), mechanisms of which remain poorly understood. Here, we demonstrate that cytoskeleton-associated protein 5 (CKAP5) serves as a cell surface target for T-DM1 and that binding of T-DM1 to CKAP5 is mediated by payload (DM1). This study introduces a novel molecular mechanism of ADC payload-mediated interaction with cell surface molecules to induce cytotoxicity. Upon binding to CKAP5, T-DM1 causes cell membrane damage and leads to calcium influx into the cells, resulting in disorganized microtubule network and apoptosis. While binding of T-DM1 with HER2 is critical for killing HER2-positive tumor cells, our data suggest that cytotoxicity induced by T-DM1 interaction with CKAP5 may preferentially damage normal cells/tissues where HER2 expression is low or missing to cause off-target toxicity. This study provides molecular basis of ADC-induced off-target cytotoxicity and opens a new avenue for developing next generation of ADCs.
Collapse
Affiliation(s)
- Yukinori Endo
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Nishant Mohan
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Yi Shen
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Jiangsong Jiang
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - David Rotstein
- Division of Compliance, Office of Surveillance and Compliance, Center for Veterinary Medicine, U.S. Food and Drug Administration (FDA), Derwood, MD, USA
| | - Wen Jin Wu
- Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD, USA
| |
Collapse
|
12
|
Challenges of Antibody Drug Conjugates in Cancer Therapy: Current Understanding of Mechanisms and Future Strategies. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s40495-018-0122-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
13
|
Mandikian D, Takahashi N, Lo AA, Li J, Eastham-Anderson J, Slaga D, Ho J, Hristopoulos M, Clark R, Totpal K, Lin K, Joseph SB, Dennis MS, Prabhu S, Junttila TT, Boswell CA. Relative Target Affinities of T-Cell-Dependent Bispecific Antibodies Determine Biodistribution in a Solid Tumor Mouse Model. Mol Cancer Ther 2018; 17:776-785. [PMID: 29339550 DOI: 10.1158/1535-7163.mct-17-0657] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/07/2017] [Accepted: 12/19/2017] [Indexed: 11/16/2022]
Abstract
Anti-HER2/CD3, a T-cell-dependent bispecific antibody (TDB) construct, induces T-cell-mediated cell death in cancer cells expressing HER2 by cross-linking tumor HER2 with CD3 on cytotoxic T cells, thereby creating a functional cytolytic synapse. TDB design is a very challenging process that requires consideration of multiple parameters. Although therapeutic antibody design strategy is commonly driven by striving for the highest attainable antigen-binding affinity, little is known about how the affinity of each TDB arm can affect the targeting ability of the other arm and the consequent distribution and efficacy. To our knowledge, no distribution studies have been published using preclinical models wherein the T-cell-targeting arm of the TDB is actively bound to T cells. We used a combined approach involving radiochemistry, invasive biodistribution, and noninvasive single-photon emission tomographic (SPECT) imaging to measure TDB distribution and catabolism in transgenic mice with human CD3ε expression on T cells. Using CD3 affinity variants, we assessed the impact of CD3 affinity on short-term pharmacokinetics, tissue distribution, and cellular uptake. Our experimental approach determined the relative effects of (i) CD3 targeting to normal tissues, (ii) HER2 targeting to HER2-expressing tumors, and (iii) relative HER2/CD3 affinity, all as critical drivers for TDB distribution. We observed a strong correlation between CD3 affinity and distribution to T-cell-rich tissues, with higher CD3 affinity reducing systemic exposure and shifting TDB distribution away from tumor to T-cell-containing tissues. These observations have important implications for clinical translation of bispecific antibodies for cancer immunotherapy. Mol Cancer Ther; 17(4); 776-85. ©2018 AACR.
Collapse
Affiliation(s)
| | | | - Amy A Lo
- Genentech, Inc., South San Francisco, California
| | - Ji Li
- Genentech, Inc., South San Francisco, California
| | | | | | - Jason Ho
- Genentech, Inc., South San Francisco, California
| | | | - Robyn Clark
- Genentech, Inc., South San Francisco, California
| | - Klara Totpal
- Genentech, Inc., South San Francisco, California
| | - Kedan Lin
- Department of Clinical Pharmacology, NGM Biopharmaceuticals Inc., South San Francisco, California
| | - Sean B Joseph
- Department of Pharmacology, Calibr, La Jolla, California
| | - Mark S Dennis
- Denali Therapeutics Inc., South San Francisco, California
| | | | | | | |
Collapse
|
14
|
Murphy PS, Patel N, McCarthy TJ. Has molecular imaging delivered to drug development? PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2017; 375:rsta.2017.0112. [PMID: 29038381 PMCID: PMC5647269 DOI: 10.1098/rsta.2017.0112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/31/2017] [Indexed: 06/07/2023]
Abstract
Pharmaceutical research and development requires a systematic interrogation of a candidate molecule through clinical studies. To ensure resources are spent on only the most promising molecules, early clinical studies must understand fundamental attributes of the drug candidate, including exposure at the target site, target binding and pharmacological response in disease. Molecular imaging has the potential to quantitatively characterize these properties in small, efficient clinical studies. Specific benefits of molecular imaging in this setting (compared to blood and tissue sampling) include non-invasiveness and the ability to survey the whole body temporally. These methods have been adopted primarily for neuroscience drug development, catalysed by the inability to access the brain compartment by other means. If we believe molecular imaging is a technology platform able to underpin clinical drug development, why is it not adopted further to enable earlier decisions? This article considers current drug development needs, progress towards integration of molecular imaging into studies, current impediments and proposed models to broaden use and increase impact.This article is part of the themed issue 'Challenges for chemistry in molecular imaging'.
Collapse
Affiliation(s)
| | - Neel Patel
- GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | | |
Collapse
|
15
|
Nejadmoghaddam MR, Zarnani AH, Ghahremanzadeh R, Ghods R, Mahmoudian J, Yousefi M, Nazari M, Ghahremani MH, Abolhasani M, Anissian A, Mahmoudi M, Dinarvand R. Placenta-specific1 (PLAC1) is a potential target for antibody-drug conjugate-based prostate cancer immunotherapy. Sci Rep 2017; 7:13373. [PMID: 29042604 PMCID: PMC5645454 DOI: 10.1038/s41598-017-13682-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/26/2017] [Indexed: 11/13/2022] Open
Abstract
Our recent findings strongly support the idea of PLAC1 being as a potential immunotherapeutic target in prostate cancer (PCa). Here, we have generated and evaluated an anti-placenta-specific1 (PLAC1)-based antibody drug conjugate (ADC) for targeted immunotherapy of PCa. Prostate cancer cells express considerable levels of PLAC1. The Anti-PLAC1 clone, 2H12C12, showed high reactivity with recombinant PLAC1 and selectivity recognized PLAC1 in prostate cancer cells but not in LS180 cells, the negative control. PLAC1 binding induced rapid internalization of the antibody within a few minutes which reached to about 50% after 15 min and almost completed within an hour. After SN38 conjugation to antibody, a drug-antibody ratio (DAR) of about 5.5 was achieved without apparent negative effect on antibody affinity to cell surface antigen. The ADC retained intrinsic antibody activity and showed enhanced and selective cytotoxicity with an IC50 of 62 nM which was about 15-fold lower compared to free drug. Anti-PLAC1-ADC induced apoptosis in human primary prostate cancer cells and prostate cell lines. No apparent cytotoxic effect was observed in in vivo animal safety experiments. Our newly developed anti-PLAC1-based ADCs might pave the way for a reliable, efficient, and novel immunotherapeutic modality for patients with PCa.
Collapse
Affiliation(s)
- Mohammad-Reza Nejadmoghaddam
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
- Immunology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran.
| | - Ramin Ghahremanzadeh
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Jafar Mahmoudian
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Yousefi
- Nanobiotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mahboobeh Nazari
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Abolhasani
- Department of Pathology, Hasheminejad Kidney Center, Iran University of Medical Sciences, IUMS, Tehran, Iran
| | - Ali Anissian
- Veterinary department, Islamic Azad University, Abhar branch, Abhar, Iran
| | - Morteza Mahmoudi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Malik P, Phipps C, Edginton A, Blay J. Pharmacokinetic Considerations for Antibody-Drug Conjugates against Cancer. Pharm Res 2017; 34:2579-2595. [PMID: 28924691 DOI: 10.1007/s11095-017-2259-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/09/2017] [Indexed: 12/26/2022]
Abstract
Antibody-drug conjugates (ADCs) are ushering in the next era of targeted therapy against cancer. An ADC for cancer therapy consists of a potent cytotoxic payload that is attached to a tumour-targeted antibody by a chemical linker, usually with an average drug-to-antibody ratio (DAR) of 3.5-4. The theory is to deliver potent cytotoxic payloads directly to tumour cells while sparing healthy cells. However, practical application has proven to be more difficult. At present there are only two ADCs approved for clinical use. Nevertheless, in the last decade there has been an explosion of options for ADC engineering to optimize target selection, Fc receptor interactions, linker, payload and more. Evaluation of these strategies requires an understanding of the mechanistic underpinnings of ADC pharmacokinetics. Development of ADCs for use in cancer further requires an understanding of tumour properties and kinetics within the tumour environment, and how the presence of cancer as a disease will impact distribution and elimination. Key pharmacokinetic considerations for the successful design and clinical application of ADCs in oncology are explored in this review, with a focus on the mechanistic determinants of distribution and elimination.
Collapse
Affiliation(s)
- Paul Malik
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada
| | - Colin Phipps
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada.,DMPK & Translational Modeling, Abbvie Inc., North Chicago, Illinois, 60064, USA
| | - Andrea Edginton
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada.
| | - Jonathan Blay
- School of Pharmacy, University of Waterloo, 10A Victoria St South, Kitchener, Ontario, N2G 1C5, Canada
| |
Collapse
|
17
|
Cilliers C, Guo H, Liao J, Christodolu N, Thurber GM. Multiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for Efficacy. AAPS JOURNAL 2016; 18:1117-1130. [PMID: 27287046 DOI: 10.1208/s12248-016-9940-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/27/2016] [Indexed: 11/30/2022]
Abstract
Antibody-drug conjugates exhibit complex pharmacokinetics due to their combination of macromolecular and small molecule properties. These issues range from systemic concerns, such as deconjugation of the small molecule drug during the long antibody circulation time or rapid clearance from nonspecific interactions, to local tumor tissue heterogeneity, cell bystander effects, and endosomal escape. Mathematical models can be used to study the impact of these processes on overall distribution in an efficient manner, and several types of models have been used to analyze varying aspects of antibody distribution including physiologically based pharmacokinetic (PBPK) models and tissue-level simulations. However, these processes are quantitative in nature and cannot be handled qualitatively in isolation. For example, free antibody from deconjugation of the small molecule will impact the distribution of conjugated antibodies within the tumor. To incorporate these effects into a unified framework, we have coupled the systemic and organ-level distribution of a PBPK model with the tissue-level detail of a distributed parameter tumor model. We used this mathematical model to analyze new experimental results on the distribution of the clinical antibody-drug conjugate Kadcyla in HER2-positive mouse xenografts. This model is able to capture the impact of the drug-antibody ratio (DAR) on tumor penetration, the net result of drug deconjugation, and the effect of using unconjugated antibody to drive ADC penetration deeper into the tumor tissue. This modeling approach will provide quantitative and mechanistic support to experimental studies trying to parse the impact of multiple mechanisms of action for these complex drugs.
Collapse
Affiliation(s)
- Cornelius Cilliers
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Hans Guo
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Jianshan Liao
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Nikolas Christodolu
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
18
|
Block M. Physiologically based pharmacokinetic and pharmacodynamic modeling in cancer drug development: status, potential and gaps. Expert Opin Drug Metab Toxicol 2016; 11:743-56. [PMID: 25940026 DOI: 10.1517/17425255.2015.1037276] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Modeling and simulation have become important means of answering questions relevant to the development of a drug, making it possible to assess risks early and to reduce costs. Physiologically based pharmacokinetic and pharmacodynamic (PBPK/PD) models contribute to a comprehensive understanding of the drug, covering specific questions from early discovery through lifecycle management stages. As for other disease areas, in oncology, PBPK and PD models are important topics that remain to be addressed. AREAS COVERED This review describes current PBPK and PD approaches, their applicability in drug development in general and specifically in the area of oncology. It discusses the current status and then focuses on key challenges and the potential for future use. It provides cases in which modeling currently cannot answer the questions and assesses the requirements to close gaps for PBPK/PD in oncology. EXPERT OPINION PBPK/PD models have led to improvements in identifying risks and reducing costs during the drug development process. Nevertheless, there is a lot of potential, where more rigorous integration of biological knowledge and specific experimental design would result in a more comprehensive biological picture. Ideally, such approaches would reveal the extent to which preclinical work can be extrapolated to clinical settings, thus enabling reliable prediction and, ultimately, reducing failed trials in clinical oncology.
Collapse
Affiliation(s)
- Michael Block
- Bayer Technology Services GmbH - Systems Pharmacology ONC , Building B106 Leverkusen , Germany
| |
Collapse
|
19
|
Akash MSH, Rehman K, Parveen A, Ibrahim M. Antibody-drug conjugates as drug carrier systems for bioactive agents. INT J POLYM MATER PO 2015. [DOI: 10.1080/00914037.2015.1038818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
20
|
Hinrichs MJM, Dixit R. Antibody Drug Conjugates: Nonclinical Safety Considerations. AAPS J 2015; 17:1055-64. [PMID: 26024656 PMCID: PMC4540738 DOI: 10.1208/s12248-015-9790-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/14/2015] [Indexed: 02/08/2023] Open
Abstract
Antibody drug conjugates (ADCs) are biopharmaceutical molecules consisting of a cytotoxic small molecule covalently linked to a targeted protein carrier via a stable cleavable or noncleavable linker. The process of conjugation yields a highly complex molecule with biochemical properties that are distinct from those of the unconjugated components. The impact of these biochemical differences on the safety and pharmacokinetic (PK) profile of the conjugate must be considered when determining the types of nonclinical safety studies required to support clinical development of ADCs. The hybrid nature of ADCs highlights the need for a science-based approach to safety assessment that incorporates relevant aspects of small and large molecule testing paradigms. This thinking is reflected in current regulatory guidelines, where sections pertaining to conjugates allow for a flexible approach to nonclinical safety testing. The aim of this article is to review regulatory expectations regarding early assessment of nonclinical safety considerations and discuss how recent advances in our understanding of ADC-mediated toxicity can be used to guide the types of nonclinical safety studies needed to support ADC clinical development. The review will also explore nonclinical testing strategies that can be used to streamline ADC development by assessing the safety and efficacy of next generation ADC constructs using a rodent screen approach.
Collapse
Affiliation(s)
- Mary Jane Masson Hinrichs
- Department of Translational Sciences, MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland, 20878, USA,
| | | |
Collapse
|
21
|
Bioanalytical approaches for characterizing catabolism of antibody–drug conjugates. Bioanalysis 2015; 7:1583-604. [DOI: 10.4155/bio.15.87] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The in vivo stability and catabolism of antibody–drug conjugates (ADCs) directly impact their PK, efficacy and safety, and metabolites of the cytotoxic or small molecule drug component of an ADC can further complicate these factors. This perspective highlights the importance of understanding ADC catabolism and the associated bioanalytical challenges. We evaluated different bioanalytical approaches to qualitatively and quantitatively characterize ADC catabolites. Here we review and discuss the rationale and experimental strategies used to design bioanalytical assays for characterization of ADC catabolism and supporting ADME studies during ADC clinical development. This review covers both large and small molecule approaches, and uses examples from Kadcyla® (T-DM1) and a THIOMAB™ antibody–drug conjugate to illustrate the process.
Collapse
|
22
|
Evolving Strategies for Target Selection for Antibody-Drug Conjugates. Pharm Res 2015; 32:3494-507. [PMID: 25585957 DOI: 10.1007/s11095-015-1624-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 01/06/2015] [Indexed: 01/06/2023]
Abstract
Antibody-drug conjugates (ADCs) represent a promising modality for the treatment of cancer. The therapeutic strategy is to deliver a potent drug preferentially to the tumor and not normal tissues by attaching the drug to an antibody that recognizes a tumor antigen. The selection of antigen targets is critical to enabling a therapeutic window for the ADC and has proven to be surprisingly complex. We surveyed the tumor and normal tissue expression profiles of the targets of ADCs currently in clinical development. Our analysis demonstrates a surprisingly broad range of expression profiles and the inability to formalize any optimal parameters for an ADC target. In this context, we discuss additional considerations for ADC target selection, including interdependencies among biophysical properties of the drug, biological functions of the target and strategies for clinical development. The TPBG (5T4) oncofetal antigen and the anti-TPBG ADC A1-mcMMAF are highlighted to demonstrate the relevance of the target's biological function. Emerging platform technologies and novel biological insights are expanding ADC target space and transforming strategies for target selection.
Collapse
|
23
|
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: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
24
|
Conner KP, Rock BM, Kwon GK, Balthasar JP, Abuqayyas L, Wienkers LC, Rock DA. Evaluation of near infrared fluorescent labeling of monoclonal antibodies as a tool for tissue distribution. Drug Metab Dispos 2014; 42:1906-13. [PMID: 25209366 PMCID: PMC11024893 DOI: 10.1124/dmd.114.060319] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/08/2014] [Indexed: 04/20/2024] Open
Abstract
The pharmacokinetic (PK) behavior of monoclonal antibodies (mAbs) is influenced by target-mediated drug disposition, off-target effects, antidrug antibody-mediated clearance, and interaction with fragment-crystallizable domain (Fc) receptors such as neonatal Fc receptor. All of these interactions hold the potential to impact mAb biodistribution. Near infrared (NIR) fluorescent probes offer an approach complementary to radionuclides to characterize drug disposition. Notably, the use of FDA-approved IRDye800 (IR800; LI-COR, Lincoln, NE) as a protein-labeling agent in preclinical work holds the potential for quantitative tissue analysis. Here, we tested the utility of the IR800 dye as a quantitative mAb tracer during pharmacokinetic analysis in both plasma and tissues using a model mouse monoclonal IgG1 (8C2) labeled with ≤1.5 molecules of IR800. The plasma PK parameters derived from a mixture of IR800-8C2 and 8C2 dosed intravenously to C57BL/6 mice at 8 mg/kg exhibited a large discrepancy in exposure depending on the method of quantitation [CLplasma = 8.4 ml/d per kilogram (NIR fluorescence detection) versus 2.5 ml/d per kilogram (enzyme-linked immunosorbent assay)]. The disagreement between measurements suggests that the PK of 8C2 is altered by addition of the IR800 dye. Additionally, direct fluorescence analysis of homogenized tissues revealed several large differences in IR800-8C2 tissue uptake when compared with a previously published study using [(125)I]8C2, most notably an over 4-fold increase in liver concentration. Finally, the utility of IR800 in combination with whole body imaging was examined by comparison of IR800-8C2 levels observed in animal sagittal cross-sections to those measured in homogenized tissues. Our results represent the first PK analysis in both mouse plasma and tissues of an IR800-mAb conjugate and suggest that mAb disposition is significantly altered by IR800 conjugation to 8C2.
Collapse
Affiliation(s)
- Kip P Conner
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Brooke M Rock
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Gayle K Kwon
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Joseph P Balthasar
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Lubna Abuqayyas
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Larry C Wienkers
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Dan A Rock
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| |
Collapse
|
25
|
New challenges and opportunities in nonclinical safety testing of biologics. Regul Toxicol Pharmacol 2014; 69:226-33. [DOI: 10.1016/j.yrtph.2014.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 04/11/2014] [Accepted: 04/12/2014] [Indexed: 12/15/2022]
|
26
|
Yip V, Palma E, Tesar DB, Mundo EE, Bumbaca D, Torres EK, Reyes NA, Shen BQ, Fielder PJ, Prabhu S, Khawli LA, Boswell CA. Quantitative cumulative biodistribution of antibodies in mice: effect of modulating binding affinity to the neonatal Fc receptor. MAbs 2014; 6:689-96. [PMID: 24572100 PMCID: PMC4011913 DOI: 10.4161/mabs.28254] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The neonatal Fc receptor (FcRn) plays an important and well-known role in antibody recycling in endothelial and hematopoietic cells and thus it influences the systemic pharmacokinetics (PK) of immunoglobulin G (IgG). However, considerably less is known about FcRn's role in the metabolism of IgG within individual tissues after intravenous administration. To elucidate the organ distribution and gain insight into the metabolism of humanized IgG1 antibodies with different binding affinities FcRn, comparative biodistribution studies in normal CD-1 mice were conducted. Here, we generated variants of herpes simplex virus glycoprotein D-specific antibody (humanized anti-gD) with increased and decreased FcRn binding affinity by genetic engineering without affecting antigen specificity. These antibodies were expressed in Chinese hamster ovary cell lines, purified and paired radiolabeled with iodine-125 and indium-111. Equal amounts of I-125-labeled and In-111-labeled antibodies were mixed and intravenously administered into mice at 5 mg/kg. This approach allowed us to measure both the real-time IgG uptake (I-125) and cumulative uptake of IgG and catabolites (In-111) in individual tissues up to 1 week post-injection. The PK and distribution of the wild-type IgG and the variant with enhanced binding for FcRn were largely similar to each other, but vastly different for the rapidly cleared low-FcRn-binding variant. Uptake in individual tissues varied across time, FcRn binding affinity, and radiolabeling method. The liver and spleen emerged as the most concentrated sites of IgG catabolism in the absence of FcRn protection. These data provide an increased understanding of FcRn's role in antibody PK and catabolism at the tissue level.
Collapse
Affiliation(s)
- Victor Yip
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Enzo Palma
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Devin B Tesar
- Drug Delivery Department; Pharma Technical Development, Genentech; South San Francisco, CA USA
| | - Eduardo E Mundo
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Daniela Bumbaca
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Elizabeth K Torres
- Non-Clinical Operations; Genentech Research & Early Development; South San Francisco, CA USA
| | - Noe A Reyes
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Ben Q Shen
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Paul J Fielder
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Saileta Prabhu
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - Leslie A Khawli
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| | - C Andrew Boswell
- Preclinical and Translational Pharmacokinetics; Genentech Research & Early Development; South San Francisco, CA USA
| |
Collapse
|
27
|
Boswell CA, Marik J, Elowson MJ, Reyes NA, Ulufatu S, Bumbaca D, Yip V, Mundo EE, Majidy N, Van Hoy M, Goriparthi SN, Trias A, Gill HS, Williams SP, Junutula JR, Fielder PJ, Khawli LA. Enhanced tumor retention of a radiohalogen label for site-specific modification of antibodies. J Med Chem 2013; 56:9418-26. [PMID: 24131491 DOI: 10.1021/jm401365h] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A known limitation of iodine radionuclides for labeling and biological tracking of receptor targeted proteins is the tendency of iodotyrosine to rapidly diffuse from cells following endocytosis and lysosomal degradation. In contrast, radiometal-chelate complexes such as indium-111-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (In-111-DOTA) accumulate within target cells due to the residualizing properties of the polar, charged metal-chelate-amino acid adduct. Iodine radionuclides boast a diversity of nuclear properties and chemical means for incorporation, prompting efforts to covalently link radioiodine with residualizing molecules. Herein, we describe the Ugi-assisted synthesis of [I-125]HIP-DOTA, a 4-hydroxy-3-iodophenyl (HIP) derivative of DOTA, and demonstration of its residualizing properties in a murine xenograft model. Overall, this study displays the power of multicomponent synthesis to yield a versatile radioactive probe for antibodies across multiple therapeutic areas with potential applications in both preclinical biodistribution studies and clinical radioimmunotherapies.
Collapse
Affiliation(s)
- C Andrew Boswell
- Genentech Research and Early Development, 1 DNA Way MS 463A , South San Francisco 94080, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Pharmacokinetic and absorption, distribution, metabolism, and excretion (ADME) characterization of antibody-drug conjugates (ADCs) reflects the dynamic interactions between the biological system and ADC, and provides critical assessments in lead selection, optimization, and clinical development. Understanding the pharmacokinetics (PK), ADME properties and consequently the pharmacokinetic-pharmacodynamic properties of ADCs is critical for their successful development. This chapter discusses the PK properties of ADCs, types of PK and ADME studies in supporting different stages of development, general design of PK/ADME studies with a focus on ADC-specific characteristics, and interpretation of PK parameters.
Collapse
|