1
|
Muccio S, Hirtz C, Kramer D, Paris J, Descloux S, Fedeli O, Deiteren A, Tribula A, Lehmann S, Vialaret J. In-depth characterization and semi-quantification of anti-drug antibodies in clinical samples using specific hybrid IC-LC-MS/MS methods. Anal Biochem 2025; 701:115797. [PMID: 39921137 DOI: 10.1016/j.ab.2025.115797] [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: 10/08/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/10/2025]
Abstract
Anti-drug antibodies (ADAs) generated by biotherapeutics can impair the drug clearance, prevent the binding to its target or lead to hypersensitivity reactions, thereby affecting efficacy and safety. It is therefore essential to assess the immunogenicity of potential biotherapeutics, particularly in clinical development. Ligand binding assays (LBA) are the gold standard for ADA detection because of their high sensitivity and throughput. However, LBA assays don't provide details on the isotypes produced and their relative abundance. As certain isotypes are known to be associated with ADA mediated adverse events, this information could be helpful to anticipate or better characterize the immunogenicity risk of biotherapeutics. A hybrid IC-LC-MS/MS strategy was developed for the detection of specific isotypes/subclasses of ADAs in a phase I clinical study. A first approach using the biotinylated drug to capture ADAs in human serum allowed the simultaneous semi-quantification of all IgG subclasses and the detection of ADAs of the IgM isotype in clinical samples. These results enabled a detailed characterization of the immune response against the biotherapeutic. A second assay was developed using a sequential immunocapture to measure drug specific IgEs known to be potentially associated with hypersensitivity reactions. The overall results were consistent with the clinical adverse events observed in some healthy volunteers.
Collapse
Affiliation(s)
- Stéphane Muccio
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France.
| | - Christophe Hirtz
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Daniel Kramer
- Sanofi, TMED-BCB, Industriepark Höchst 65926 Frankfurt, Germany
| | - Johanna Paris
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Sandrine Descloux
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Olivier Fedeli
- Sanofi, TMED-BCB, 371 rue du Professeur Blayac, 34184 Montpellier, France
| | - Annemie Deiteren
- Ablynx R&D, TMED-TMCP, Technologiepark-Zwijnaarde 21, 9052 Gent/Zwijnaarde, Belgium
| | | | - Sylvain Lehmann
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France.
| | - Jérôme Vialaret
- Montpellier Univ, IRMB CHU, INM INSERM, 80 avenue Augustin Fliche, 34295 Montpellier, France
| |
Collapse
|
2
|
Chen S, Piatkov K, Dong L, Sugimoto H. Detection of antidrug antibodies against antibody-drug conjugates by solid-phase extraction with acid dissociation in cynomolgus monkey serum. Drug Metab Dispos 2025; 53:100039. [PMID: 40037092 DOI: 10.1016/j.dmd.2025.100039] [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: 10/10/2024] [Accepted: 01/14/2025] [Indexed: 03/06/2025] Open
Abstract
The formation of antidrug antibodies (ADAs) against antibody-drug conjugates (ADCs) can trigger a humoral immune response and change drug exposure. Although the immunogenicity assessment of an ADC drug in nonclinical nonhuman primates may not directly translate to potential immunogenicity in humans, the nonclinical ADA assay facilitates understanding the pharmacokinetic profiles of biotherapeutics. The immune response against the human IgG4 monoclonal antibody-based ADC was suspected in cynomolgus monkey serum after intravenous administration at 1.5 mg/kg. However, the conventional bridging format ADA assay presented unique challenges for the ADC molecules due to the interaction of ADC-based capture and detection reagents, which generated high background noise. Solid-phase extraction with acid dissociation (SPEAD) sample treatment allowed the selective ADA transfer to a second plate for detection while avoiding the interaction between the capture and detection reagents. The signal-to-noise ratio in the ADA assay for ADCs was notably improved with SPEAD sample treatment compared with the results from the bridging assay. Importantly, the rapid drug clearance of the ADC molecules at the later time points was well correlated with the signal-to-noise ratio of the ADA assay in monkey serum, suggesting the validity of the results. Hence, we demonstrated the utility of the SPEAD sample treatment to mitigate the critical reagent interaction that triggered the unexpectedly high background in the ADA assay. SIGNIFICANCE STATEMENT: A fit-for-purpose antidrug antibody screening assay for the human IgG4 monoclonal antibody-based antibody-drug conjugate (ADC) molecule by solid-phase extraction with acid dissociation was developed to mitigate the high background noise due to the interaction of capture and detection ADCs. A positive antidrug antibody signal was observed in the monkey serum sample, which is in line with the significant decrease in the plasma concentration of ADCs at the later time points.
Collapse
Affiliation(s)
- Susan Chen
- Drug Metabolism and Pharmacokinetics and Modeling (DMPK&M), Takeda Development Center Americas Inc, Cambridge, Massachusetts
| | - Konstantin Piatkov
- Drug Metabolism and Pharmacokinetics and Modeling (DMPK&M), Takeda Development Center Americas Inc, Cambridge, Massachusetts
| | - Linlin Dong
- Drug Metabolism and Pharmacokinetics and Modeling (DMPK&M), Takeda Development Center Americas Inc, Cambridge, Massachusetts
| | - Hiroshi Sugimoto
- Drug Metabolism and Pharmacokinetics and Modeling (DMPK&M), Takeda Development Center Americas Inc, Cambridge, Massachusetts.
| |
Collapse
|
3
|
Thway TM, Aluri J, Lalovic B, Ooi CE, Sauter N, Xing D, Czuczman M, Yasuda S. Effect of E7777 Immunogenicity on Pharmacokinetics, Efficacy, and Safety in Adult Patients With Relapsed or Refractory Cutaneous T-Cell Lymphoma. Clin Transl Sci 2025; 18:e70166. [PMID: 40009571 PMCID: PMC11864228 DOI: 10.1111/cts.70166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/23/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
E7777 is a therapeutic recombinant fusion protein comprised of diphtheria toxin fragments and human interleukin-2 (IL-2). Treatment with E7777 generates anti-drug antibodies (ADA). E7777-G000-302 assessed the efficacy and safety of E7777 in patients with relapsed or refractory cutaneous T-cell lymphoma (CTCL). Here, we describe the association between E7777 (at 9 μg/kg) and ADAs, and its effect on pharmacokinetics (PK), efficacy, and safety. Of 91 patients with immunogenicity results at baseline, 78.0% and 5.5% had preexisting anti-E7777 (E-ADAs) and anti-IL-2 antibodies (I-ADAs), respectively. The prevalence of E-ADAs and I-ADAs peaked at C3D1 (median titer 650,175) and C8D1 (median titer 32,805), respectively. However, I-ADA fold increases (1312-fold) were higher than E-ADA increases (181-fold). ADAs' effect on PK was assessed after treatment. E7777 reached Cmax at 60 min after infusion, with clearance of 44.6 mL/min at C1D1 and 133 mL/min at C5D1. Cmax and AUC(0-t) decreased by 60% and 84%, respectively, at C5D1 compared with C1D1. Of patients in the safety analysis set (n = 84) evaluated for treatment-emergent (TE) ADAs, 92.9% were considered TE E-ADA positive and 82.1% were TE I-ADA positive. The objective response rate was 39.0% in TE E-ADA-positive patients and 42.6% in TE I-ADA-positive patients. Serious TE adverse events were reported by 32.1% of TE E-ADA-positive patients, 29.0% of TE I-ADA-positive patients, 100% of E-ADA-negative patients, and 73.3% of I-ADA-negative patients. These results indicate that the presence of ADAs decreased E7777 exposure over time but did not adversely impact efficacy and safety in patients with CTCL.
Collapse
Affiliation(s)
| | - Jagadeesh Aluri
- Clinical Pharmacology Science, Eisai Inc.NutleyNew JerseyUSA
| | | | - Chean E. Ooi
- Clinical DevelopmentOncology Eisai Inc.NutleyNew JerseyUSA
| | | | - Dongyuan Xing
- Clinical DevelopmentOncology Eisai Inc.NutleyNew JerseyUSA
| | - Myron Czuczman
- Citius Oncology Pharmaceuticals Inc.CranfordNew JerseyUSA
| | - Sanae Yasuda
- Clinical Pharmacology Science, Eisai Inc.NutleyNew JerseyUSA
| |
Collapse
|
4
|
Sirohi P, Hainline K, Bullock H, Wang S, Konrad RJ, Wen Y. Stability of Anti-Drug Antibodies Against Antibody Therapeutics. AAPS J 2025; 27:45. [PMID: 39953303 DOI: 10.1208/s12248-025-01030-z] [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: 11/06/2024] [Accepted: 01/24/2025] [Indexed: 02/17/2025] Open
Abstract
Biotherapeutics have the potential to elicit unwanted immune responses that can lead to the production of anti-drug antibodies (ADA). It is critical that ADA responses are detected, characterized, and monitored to understand the safety and efficacy of a drug. ADA samples must remain stable in long- and short-term storage conditions to ensure reliable analysis. Whereas the stability of anti-vaccine antibodies has been well-studied, there are few reports examining the stability of anti-therapeutic antibodies using clinical samples. In this study, ADA samples from four clinical trials of antibody therapeutics were found to be stable after long-term storage (1-10 years) at -80°C and short-term storage (24 h to two weeks) at 4°C, 22°C, and 37°C. In addition, samples were stable after 16 freeze/thaw cycles. The results demonstrate the stability of ADA in clinical samples under various conditions. Consequently, the results observed herein suggest that the routine assessment of ADA sample stability may not be warranted.
Collapse
Affiliation(s)
- Parul Sirohi
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, U.S.A
| | - Kelly Hainline
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, U.S.A..
| | - Heather Bullock
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, U.S.A
| | - Sihe Wang
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, U.S.A
| | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, U.S.A
| | - Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, U.S.A
| |
Collapse
|
5
|
Cohen JR, Joubert MK, Tabassum S, Capili A, Carreon J, Xiang C, Prabhu S, Merlo A, Mytych D, Dolan DG, Kouda R. Experimental validation of a parenteral permitted daily exposure value for cleaning-induced degradants from recombinant therapeutic proteins with in vitro immunogenicity assays. J Pharm Sci 2025; 114:866-877. [PMID: 39490658 DOI: 10.1016/j.xphs.2024.10.041] [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: 06/21/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Multiproduct manufacturing of biotherapeutic proteins generate cleaning-induced protein degradants because of extreme pH and temperature conditions during the cleaning process. Cleaning Acceptance limits are calculated based on the maximum allowable carryover (MAC) assessment of the previously manufactured active pharmaceutical ingredient (API) - or drug product - based on the permitted daily exposure (PDE) of the previously manufactured API into the dose of subsequent product. In this study, we tested a previously determined PDE value for cleaning-induced protein degradants of 650 µg/dose. A bench-scale cleaning method was used to generate cleaning induced degradants from both a half-life extension (HLE) BiTE® molecule and a mAb product. For this investigation, degradants of HLE BiTE®-A and mAb-1 were characterized alone or after spiking of 650 µg of degradants of HLE BiTE®-A or 650 µg degradants of mAb-1, into mAb-1, respectively. These samples were characterized by endotoxin testing, size exclusion chromatography (SEC), light obscuration by HIAC, and micro-fluidic imaging (MFI). These results suggest that significant degradation of the molecule occurs because of the cleaning procedure, and it is no longer in the intact form or active state. The potential immogenic impact was assessed using a cell line assay to assess immune activation, and a human Peripheral Blood Mononuclear Cell (PBMC) assay to assess T cell activation, T cell proliferation, and cytokine release after 20 h and 7 days. Findings from the various in vitro cell-based immune activation assays suggest that the presence of 650 µg of carryover of degradants either alone or spiked into the same or a cross-product do not increase immunogenicity risk in cell-based assays - suggesting that the current PDE of 650 µg/dose for cleaning-induced degradant carryover does not have a risk of immunogenicity in patients.
Collapse
Affiliation(s)
- Joseph R Cohen
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States.
| | - Marisa K Joubert
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Syeda Tabassum
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Allyson Capili
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Julia Carreon
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Cathie Xiang
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Siddharth Prabhu
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Anthony Merlo
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States
| | - Dan Mytych
- The Department of Clinical Immunology, Amgen Inc., Thousand Oaks, CA 91320, United States
| | - David G Dolan
- The Department of Environmental Health and Safety, Amgen Inc., Thousand Oaks, CA 91320, United States
| | - Ram Kouda
- The Department of Process Development, Amgen Inc., One Amgen Center Dr., Thousand Oaks, CA 91320, United States.
| |
Collapse
|
6
|
Genaro LM, Carron J, de Castro MM, Franceschini APMDF, Lourenço GJ, da Cruz CKNV, Reis GFSR, Pascoal LB, Mello JDC, Pereira IM, Nascimento ML, Oliveira PDSP, Corona LP, Ayrizono MDLS, Lima CSP, Leal RF. Therapeutic drug monitoring and immunogenetic factors associated with the use of adalimumab in Crohn's disease patients. Int J Immunopathol Pharmacol 2025; 39:3946320251319379. [PMID: 39959979 PMCID: PMC11831650 DOI: 10.1177/03946320251319379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 01/24/2025] [Indexed: 02/20/2025] Open
Abstract
Crohn's disease (CD) involves immune system interactions with intestinal tissue, driven by pro-inflammatory cytokines like Tumor Necrosis Factor (TNF-α). Adalimumab, targeting TNF-α, regulates associated inflammatory responses. Despite being humanized, it may induce immunogenic processes, affecting treatment effectiveness. Thus, monitoring serum adalimumab and anti-drug antibody (ADA) levels can optimize therapy. Understanding genetic factors influencing adalimumab response can enhance personalized treatment and improve patient quality of life. We aimed to quantify adalimumab serum levels, assess test interchangeability, detect ADA, examine immune complex formation, and investigate genetic phenotypes related to immunogenicity in CD patients. Seventy CD patients in the maintenance phase with adalimumab were classified into active (CDA) and remission (CDR) groups. Adalimumab concentration was determined via enzyme-linked immunosorbent assay (ELISA-Promonitor) and lateral flow assay (Quantum Blue), with assay interchangeability assessed statistically. ADA and immune complex formation were quantified using ELISA assays. DNA was genotyped for the genes ATG16L1, CD96, and CD155. No significant differences in adalimumab serum concentrations were observed between groups, regardless of the assay. However, a statistical difference between the tests indicated measurement disparity (P = 0.003), with moderate agreement (Lin's correlation of 0.247). ADA was detected in 4 of 27 of the patients with infratherapeutic levels, 3 in the CDA group and 1 in the CDR group. Analysis of immune complexes revealed significantly higher concentrations in the CDA group (P = 0.0125). The genotypic evaluation revealed significant associations for the CD96 CC (wild-type) genotype with higher CRP levels, colonic involvement, and infratherapeutic levels of adalimumab. ATG16L1 CC genotype was associated with higher CDEIS and fecal calprotectin values, while the variant (TT) genotype had lower platelet counts. The effectiveness of treatment with adalimumab was not directly related to higher medication levels in this cohort. The disparity between tests indicates the need to use only one test in patient follow-up to ensure accuracy in therapeutic monitoring. Genotypic differences highlight the correlation between the wild genotype for CD96 and ATG16L1 with unfavorable laboratory and endoscopic response to adalimumab. Finally, the more significant levels of immune complexes in the CDA group indicate an association with a worse response to adalimumab.
Collapse
Affiliation(s)
- Livia Moreira Genaro
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Juliana Carron
- Laboratory of Cancer Genetics (Lageca), School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Marina Moreira de Castro
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Ana Paula Menezes de Freitas Franceschini
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Gustavo Jacob Lourenço
- Laboratory of Cancer Genetics (Lageca), School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | | | | | - Livia Bitencourt Pascoal
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Juliana Delgado Campos Mello
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Isabela Machado Pereira
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Millene Leal Nascimento
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Priscilla De Sene Portel Oliveira
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Ligiana Pires Corona
- Nutritional Epidemiology Laboratory, School of Applied Sciences, University of Campinas (Unicamp), Limeira, São Paulo, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Carmen Silvia Passos Lima
- Laboratory of Cancer Genetics (Lageca), School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| | - Raquel Franco Leal
- Inflammatory Bowel Disease Research Laboratory (LabDII), Gastrocenter, Colorectal Surgery Unit, Surgery Department, School of Medical Sciences, University of Campinas (Unicamp), Campinas, São Paulo, Brazil
| |
Collapse
|
7
|
Swanson SJ. What are clinically significant anti-drug antibodies and why is it important to identify them. Front Immunol 2024; 15:1401178. [PMID: 39737166 PMCID: PMC11682980 DOI: 10.3389/fimmu.2024.1401178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/22/2024] [Indexed: 01/01/2025] Open
Abstract
The FDA has released new draft guidance to standardize how immunogenicity of protein therapeutics is described in product labels. A key aspect to this new guidance is that companies should describe anti-drug antibodies that have clinical significance in addition to reporting ADAs' incidence. Factors to consider when determining clinical significance include if those antibodies have a significant effect on the drug's pharmacokinetics, pharmacodynamics, efficacy, and/or safety. While in many instances, the humoral response to protein therapeutics does not have any clinical significance, there are cases where there is a clinically significant effect and it is important to communicate this information to physicians and patients. This new guidance also delineates where immunogenicity information should be listed in product labels which should provide consistency in how this information is listed. There are many factors that contribute to a therapeutic's immunogenicity and determining clinical significance is both complex and challenging, requiring that companies perform thorough analyses with scientific rigor. The analysis that is now proposed to understand clinical significance of ADAs is a new concept and will require companies to develop a strategy for compliance. This manuscript sets forth some of the key considerations in answering this important question. One of the benefits that this new guidance will provide is a common approach for describing the immune response to therapeutics that will be located in a dedicated section of the label, providing valuable consistency across protein therapeutics. Section 12.6 in the Clinical Pharmacology portion of the label will contain the relevant immunogenicity information, which will make it much simpler to find immunogenicity information in product labels. This new guidance is currently being utilized for new protein therapeutics and companies are being requested to systematically revise the labels of previously approved drugs for compliance, although an absolute timeline for this has not been established as of this writing.
Collapse
Affiliation(s)
- Steven James Swanson
- translational Pharmacokinetics Pharmacodynamics (tPKPD), Genentech Inc., San Francisco, CA, United States
| |
Collapse
|
8
|
Liu W, Yang J, Yan W, Peng K. Reformation of a Clinical Anti-Drug Antibody Assay to Enable the Immunogenicity Assessment of a Bispecific Antibody Biotherapeutic. AAPS J 2024; 27:12. [PMID: 39663290 DOI: 10.1208/s12248-024-00996-6] [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: 09/05/2024] [Accepted: 11/09/2024] [Indexed: 12/13/2024] Open
Abstract
An enzyme-linked immunosorbent assay (ELISA) based anti-drug antibody (ADA) assay was developed to support the clinical development of a bispecific antibody biotherapeutic anti-A/B. This anti-A/B clinical ADA Version 1 (V1) assay was successfully validated initially using commercial samples from the target indication. However, applying the validation cut point factors (CPFs) led to a high untreated ADA positive rate in the Phase 1 study baseline sample analysis. While implementing the in-study CPFs was effective to mitigate the high baseline prevalence, this led to unfavorable assay sensitivity with no drug tolerance, which necessitated an assay re-optimization. The re-optimized Version 2 assay (V2) was able to mitigate the matrix interference observed in the clinical sample testing using the V1 assay, proven to be a more suitable method. The V2 assay optimization work was discussed, and the performance of the V1 and V2 assays during validation and clinical sample analysis was compared. Preliminary sample testing results generated using the two versions of the assay were compared and the ADA clinical impact was discussed. Our experience insinuates that a successfully validated method does not guarantee to be appropriate for sample testing. Adjustments of the method may be required to ensure that it performs as expected during sample testing and throughout the assay's lifecycle. This work highlights the importance of verifying the assay suitability during clinical sample testing and making appropriate adjustments as needed, especially in the first clinical study and the first study for a new indication.
Collapse
Affiliation(s)
- Wenyu Liu
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA
| | - Jie Yang
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA
| | - Weili Yan
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA
| | - Kun Peng
- Department of BioAnalytical Sciences, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080-4990, USA.
| |
Collapse
|
9
|
Leisegang R, Silber Baumann HE, Lennon-Chrimes S, Ito H, Miya K, Genin JC, Plan EL. Immunogenicity dynamics and covariate effects after satralizumab administration predicted with a hidden Markov model. CPT Pharmacometrics Syst Pharmacol 2024; 13:2171-2184. [PMID: 39380259 DOI: 10.1002/psp4.13230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 10/10/2024] Open
Abstract
Immunogenicity is the propensity of a therapeutic protein to generate an immune response to itself. While reporting of antidrug antibodies (ADAs) is increasing, model-based analysis of such data is seldom performed. Model-based characterization of factors affecting the emergence and dissipation of ADAs may inform drug development and/or improve understanding in clinical practice. This analysis aimed to predict ADA dynamics, including the potential influence of individual covariates, following subcutaneous satralizumab administration. Satralizumab is a humanized IgG2 monoclonal recycling IL-6 receptor antagonist antibody approved for treating neuromyelitis optica spectrum disorder (NMOSD). Longitudinal pharmacokinetic (PK) and ADA data from 154 NMOSD patients in two pivotal Phase 3 studies (NCT02028884, NCT02073279) and PK data from one Phase 1 study (SA-001JP) in 72 healthy volunteers were available for this analysis. An existing population PK model was adapted to derive steady-state concentration without ADA for each patient. A mixed hidden Markov model (mHMM) was developed whereby three different states were identified: one absorbing Markov state for non-ADA developer, and two dynamic and inter-connected Markov states-transient ADA negative and positive. Satralizumab exposure and body mass index impacted transition probabilities and, therefore, the likelihood of developing ADAs. In conclusion, the mHMM model was able to describe the time course of ADA development and identify patterns of ADA development in NMOSD patients following treatment with satralizumab, which may allow for the formulation of strategies to reduce the emergence or limit the impact of ADA in the clinical setting.
Collapse
Affiliation(s)
- Rory Leisegang
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hanna E Silber Baumann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | | | | | | | - Jean-Christophe Genin
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Elodie L Plan
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Hagman C, Chasseigne G, Nelson R, Anlauff F, Kagan M, Goldfine AB, Terszowski G, Jadhav M. Immunogenicity assessment strategy for a chemically modified therapeutic protein in clinical development. Front Immunol 2024; 15:1438251. [PMID: 39588370 PMCID: PMC11586355 DOI: 10.3389/fimmu.2024.1438251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 10/16/2024] [Indexed: 11/27/2024] Open
Abstract
The clinical immunogenicity assessment for complex multidomain biological drugs is challenging due to multiple factors that must be taken into consideration. Here, we describe a strategy to overcome multiple bioanalytical challenges in order to assess anti-drug antibodies (ADA) for a novel and unique chemically modified protein therapeutic. A risk-centered approach was adopted to evaluate the immunogenic response to a modified version of human growth differentiation factor 15 (GDF15) connected to an albumin-binding fatty acid via a polyethylene glycol (PEG) linker. Key steps include monitoring anti-drug antibodies (ADAs), using a standard tiered approach of screening and confirmation. To deepen our understanding of ADA response, as a third tier of immunogenicity assessment, novel extensive characterization using a set of assays was developed, validated, and used routinely in clinical sample analysis. This characterization step included performance of titration, mapping of ADA response including anti-GDF15 and anti-PEG-fatty-acid antibody characterization, and assessment of the neutralizing anti-drug antibodies (NAbs) using cell-based assays for immunogenicity in parallel. The analytical methods were applied during two clinical trials involving both healthy volunteers and overweight or obese patients. We observed low incident rates for ADA and no ADAs against the PEG linker with fatty acid conjugation. In one of the clinical studies, we identified neutralizing ADAs. The proposed novel strategy of extensive characterization proved effective for monitoring the presence of ADAs and NAbs and can be used to support clinical development of a broad range of chemically modified proteins and multidomain biotherapeutics.
Collapse
Affiliation(s)
- Charlotte Hagman
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Basel, Switzerland
| | - Gaetan Chasseigne
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Basel, Switzerland
| | | | | | - Mark Kagan
- Pharmacokinetic Sciences, Biomedical Research, Novartis, East Hanover, NJ, United States
| | - Allison B. Goldfine
- Translational Medicine, Biomedical Research, Novartis, Cambridge, MA, United States
| | - Grzegorz Terszowski
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Basel, Switzerland
| | - Maria Jadhav
- Pharmacokinetic Sciences - Drug Disposition, Biomedical Research, Novartis, Cambridge, MA, United States
| |
Collapse
|
11
|
Choi SM, Lee JH, Ko S, Hong SS, Jin HE. Mechanism of Action and Pharmacokinetics of Approved Bispecific Antibodies. Biomol Ther (Seoul) 2024; 32:708-722. [PMID: 39448393 PMCID: PMC11535297 DOI: 10.4062/biomolther.2024.146] [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: 08/21/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Bispecific antibodies represent a significant advancement in therapeutic antibody engineering, offering the ability to simultaneously target two distinct antigens. This dual-targeting capability enhances therapeutic efficacy, especially in complex diseases, such as cancer and autoimmune disorders, where drug resistance and incomplete target coverage are prevalent challenges. Bispecific antibodies facilitate immune cell engagement and disrupt multiple signaling pathways, providing a more comprehensive treatment approach than traditional monoclonal antibodies. However, the intricate structure of bispecific antibodies introduces unique pharmacokinetic challenges, including issues related to their absorption, distribution, metabolism, and excretion, which can significantly affect their efficacy and safety. This review provides an in-depth analysis of the structural design, mechanisms of action, and pharmacokinetics of the currently approved bispecific antibodies. It also highlights the engineering innovations that have been implemented to overcome these challenges, such as Fc modifications and advanced dimerization techniques, which enhance the stability and half-life of bispecific antibodies. Significant progress has been made in bispecific antibody technology; however, further research is necessary to broaden their clinical applications, enhance their safety profiles, and optimize their incorporation into combination therapies. Continuous advancements in this field are expected to enable bispecific antibodies to provide more precise and effective therapeutic strategies for a range of complex diseases, ultimately improving patient outcomes and advancing precision medicine.
Collapse
Affiliation(s)
- Seong Min Choi
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Ju-Hee Lee
- College of Korean Medicine, Dongguk University, Goyang 10326, Republic of Korea
| | - Soyeon Ko
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine & Program in Biomedicals Science & Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Hyo-Eon Jin
- Department of Biohealth Regulatory Science, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
12
|
Jacquot G, Lopez Navarro P, Grange C, Boudali L, Harlepp S, Pivot X, Detappe A. Landscape of Subcutaneous Administration Strategies for Monoclonal Antibodies in Oncology. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406604. [PMID: 39165046 DOI: 10.1002/adma.202406604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/15/2024] [Indexed: 08/22/2024]
Abstract
In recent decades, subcutaneous (SC) administration of monoclonal antibodies (mAbs) has emerged as a promising alternative to intravenous delivery in oncology, offering comparable therapeutic efficacy while addressing patient preferences. This perspective article provides an in-depth analysis of the technological landscape surrounding SC mAb administration in oncology. It outlines various technologies under evaluation across developmental stages, spanning from preclinical investigations to the integration of established methodologies in clinical practice. Additionally, this perspective article explores emerging trends and prospective trajectories, shedding light on the evolving landscape of SC mAb administration. Furthermore, it emphasizes key checkpoints related to quality attributes essential for optimizing mAb delivery via the SC route. This review serves as a valuable resource for researchers, clinicians, and healthcare policymakers, offering insights into the advancement of SC mAb administration in oncology and its implications for patient care.
Collapse
Affiliation(s)
- Guillaume Jacquot
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Pedro Lopez Navarro
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Coralie Grange
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Lotfi Boudali
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Sébastien Harlepp
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe, Strasbourg, 67000, France
- Equipe labellisée Ligue contre le Cancer, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, 67000, France
- Equipe de Synthèse Pour l'Analyse, Institut Pluridisciplinaire Hubert Curien (IPHC), UMR 7178 CNRS/University of Strasbourg, Strasbourg, Cedex 2, 67087, France
| |
Collapse
|
13
|
Masuda T, Funakoshi T, Horimatsu T, Yamamoto S, Matsubara T, Masui S, Nakagawa S, Ikemi Y, Yanagita M, Muto M, Terada T, Yonezawa A. Low serum concentrations of bevacizumab and nivolumab owing to excessive urinary loss in patients with proteinuria: a case series. Cancer Chemother Pharmacol 2024; 94:615-622. [PMID: 38456954 DOI: 10.1007/s00280-024-04659-3] [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: 12/26/2023] [Accepted: 02/18/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE Proteinuria can cause interindividual variability in the pharmacokinetics of therapeutic antibodies and may affect therapeutic efficacy. Here, we measured the serum and urinary concentrations of bevacizumab (BV) and nivolumab (NIVO) in patients with proteinuria and reported a case series of these patients. METHODS Thirty-two cancer patients who received BV every 3 weeks or NIVO every 2 weeks between November 2020 and September 2021 at Kyoto University Hospital were enrolled in this study. The serum and urinary concentrations of BV and NIVO were measured using liquid chromatography-tandem mass spectrometry. RESULTS We divided the BV-treated patients and the NIVO-treated patients into two groups based on the urine protein-creatinine ratio (UPCR): UPCR 1 g/g or higher (BV, n = 9; NIVO, n = 3) and UPCR less than 1 g/g (BV, n = 14; NIVO, n = 6). Serum concentrations of the therapeutic antibodies adjusted by their doses were significantly lower in both BV- and NIVO-treated patients with UPCR 1 g/g or higher compared to those with less than 1 g/g. In patients with UPCR 1 g/g or higher, urinary concentrations of the therapeutic antibodies adjusted by their serum concentrations and urinary creatinine concentrations tended to increase. CONCLUSION This case-series study suggests a possibility of reduction in serum concentrations of BV and NIVO in patients with proteinuria by urinary excretion of these drugs.
Collapse
Affiliation(s)
- Takashi Masuda
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Taro Funakoshi
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Horimatsu
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Yamamoto
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Matsubara
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sho Masui
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato- ku, Tokyo, 105-8512, Japan
| | - Shunsaku Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Yasuaki Ikemi
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Terada
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Atsushi Yonezawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan.
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato- ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
14
|
Herrera M, Pretelli G, Desai J, Garralda E, Siu LL, Steiner TM, Au L. Bispecific antibodies: advancing precision oncology. Trends Cancer 2024; 10:893-919. [PMID: 39214782 DOI: 10.1016/j.trecan.2024.07.002] [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: 04/01/2024] [Revised: 06/29/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Bispecific antibodies (bsAbs) are engineered molecules designed to target two different epitopes or antigens. The mechanism of action is determined by the bsAb molecular targets and structure (or format), which can be manipulated to create variable and novel functionalities, including linking immune cells with tumor cells, or dual signaling pathway blockade. Several bsAbs have already changed the treatment landscape of hematological malignancies and select solid cancers. However, the mechanisms of resistance to these agents are understudied and the management of toxicities remains challenging. Herein, we review the principles in bsAb engineering, current understanding of mechanisms of action and resistance, data for clinical application, and provide a perspective on ongoing challenges and future developments in this field.
Collapse
Affiliation(s)
- Mercedes Herrera
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Giulia Pretelli
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jayesh Desai
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Department of Medical Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Lillian L Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Thiago M Steiner
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lewis Au
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
| |
Collapse
|
15
|
Ayoun Alsoud R, Le Moan N, Holten-Andersen L, Knudsen T, Lennernäs H, Simonsson USH. Model-Based Interspecies Scaling for Predicting Human Pharmacokinetics of CB 4332, a Complement Factor I Protein. J Pharm Sci 2024; 113:2895-2903. [PMID: 38945365 DOI: 10.1016/j.xphs.2024.06.022] [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: 03/04/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
Interspecies scaling of the pharmacokinetics (PK) of CB 4332, a 150 kDa recombinant complement factor I protein, was performed using traditional and model-based approaches to inform first-in-human dose selection. Plasma concentration versus time data from four preclinical PK studies of single intravenous and subcutaneous (SC) CB 4332 dosing in mice, rats and nonhuman primates (NHPs) were modeled simultaneously using naive pooling including allometric scaling. The human-equivalent dose was calculated using the preclinical no observed adverse effect level (NOAEL) as part of the dose-by-factor approach. Pharmacokinetic modeling of CB 4332 revealed species-specific differences in the elimination, which was accounted for by including an additional rat-specific clearance. Signs of anti-drug antibodies (ADA) formation in all rats and some NHPs were observed. Consequently, an additional ADA-induced clearance parameter was estimated including the time of onset. The traditional dose-by-factor approach calculated a maximum recommended starting SC dose of 0.9 mg/kg once weekly, which was predicted it to result in a trough steady-state concentration lower than the determined efficacy target range for CB 4332 in humans. Model simulations predicted the efficacy target range to be reached using 5 mg/kg once weekly SC dosing.
Collapse
Affiliation(s)
- Rami Ayoun Alsoud
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | - Tom Knudsen
- Catalyst Biosciences, South San Francisco, California, USA
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
16
|
Hartinger JM, Šíma M, Hrušková Z, Pilková A, Krátký V, Ryšavá R, Jančová E, Bobek D, Douša J, Francová I, Tesař V, Slanař O. A novel dosing approach for rituximab in glomerular diseases based on a population pharmacokinetic analysis. Biomed Pharmacother 2024; 175:116655. [PMID: 38678967 DOI: 10.1016/j.biopha.2024.116655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024] Open
Abstract
OBJECTIVES Rituximab is being increasingly prescribed for the treatment of autoimmune glomerular diseases. While it is highly effective for some diseases, the response is less predictable for others, which may be due to differing requirements in terms of the dosing according to the disease type and variations concerning exposure to the drug. METHODS We compiled novel rituximab dosing schedules according to pharmacokinetic analysis of data gathered from rituximab treated patients in a tertiary referral nephrology centre between May 2020 and June 2023. The population-pharmacokinetic analysis was based on the rituximab dosing, the patients' characteristics, rituximab levels and anti-rituximab antibodies. RESULTS The analysis, which was based on data from 185 patients, clearly highlighted differing rituximab dosing requirements for patients with ANCA associated vasculitis and minimal change disease compared to those with membranous nephropathy, focal-segmental glomerulosclerosis and lupus nephritis. This corresponded to the good treatment response of the first two diseases and the unreliable efficacy for the others. The model predicts the rituximab pharmacokinetics with high degree of accuracy when body weight, proteinuria, type of glomerulonephritis, treatment length and anti-rituximab antibodies formation are used as covariates. We proposed a dosing schedule with shortened dosing intervals for difficult-to-treat diagnoses with high proteinuria. CONCLUSION In order to ensure reliable and comparable exposure of rituximab with respect to the full range of glomerular diseases, the dosing schedule should be adjusted for membranous nephropathy, focal-segmental glomerulosclerosis and lupus nephritis. This is largely, but not solely, due to the enhanced level of unselective proteinuria in these diseases.
Collapse
Affiliation(s)
- Jan Miroslav Hartinger
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.
| | - Martin Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Zdenka Hrušková
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Alena Pilková
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Vojtěch Krátký
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Romana Ryšavá
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Eva Jančová
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Daniel Bobek
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jiří Douša
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ivana Francová
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Vladimír Tesař
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.
| |
Collapse
|
17
|
Tiwari P, Yadav K, Shukla RP, Bakshi AK, Panwar D, Das S, Mishra PR. Extracellular vesicles-powered immunotherapy: Unleashing the potential for safer and more effective cancer treatment. Arch Biochem Biophys 2024; 756:110022. [PMID: 38697343 DOI: 10.1016/j.abb.2024.110022] [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: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Cancer treatment has seen significant advancements with the introduction of Onco-immunotherapies (OIMTs). Although some of these therapies have received approval for use, others are either undergoing testing or are still in the early stages of development. Challenges persist in making immunotherapy widely applicable to cancer treatment. To maximize the benefits of immunotherapy and minimize potential side effects, it's essential to improve response rates across different immunotherapy methods. A promising development in this area is the use of extracellular vesicles (EVs) as novel delivery systems. These small vesicles can effectively deliver immunotherapies, enhancing their effectiveness and reducing harmful side effects. This article discusses the importance of integrating nanomedicines into OIMTs, highlighting the challenges with current anti-OIMT methods. It also explores key considerations for designing nanomedicines tailored for OIMTs, aiming to improve upon existing immunotherapy techniques. Additionally, the article looks into innovative approaches like biomimicry and the use of natural biomaterial-based nanocarriers (NCs). These advancements have the potential to transform the delivery of immunotherapy. Lastly, the article addresses the challenges of moving OIMTs from theory to clinical practice, providing insights into the future of using advanced nanotechnology in cancer treatment.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Sweety Das
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
18
|
Lotz GP, Lutz A, Martin-Facklam M, Hansbauer A, Schick E, Moessner E, Antony M, Stuchly T, Viert M, Hosse RJ, Freimoser-Grundschober A, Klein C, Schäfer M, Ritter M, Stubenrauch KG. Characterization of anti-drug antibody responses to the T-cell engaging bispecific antibody cibisatamab to understand the impact on exposure. Front Immunol 2024; 15:1406353. [PMID: 38881900 PMCID: PMC11176492 DOI: 10.3389/fimmu.2024.1406353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
An appropriately designed pharmacokinetic (PK) assay that is sensitive for anti-drug antibody (ADA) impact on relevant exposure is an alternative strategy to understand the neutralizing potential of ADAs. However, guidance on how to develop such PK assays and how to confirm the functional ADA impact on exposure is missing. Here, the PK assay of a T-cell-engaging bispecific antibody, cibisatamab, was developed based on its mechanism of action (MoA). Using critical monoclonal anti-idiotypic (anti-ID) antibody positive controls as ADA surrogates, the impact on exposure was evaluated pre-clinically. In a phase I clinical trial (NCT02324257), initial data suggest that the combination of ADA and PK assays for correlation of the ADA response with cibisatamab exposure. To understand the neutralizing potential of patient-derived ADAs on drug activity, advanced ADA characterization has been performed. Structural binding analysis of ADAs to antibody domains of the drug and its impact on targeting were assessed. For this purpose, relevant patient ADA binding features were identified and compared with the specific monoclonal anti-ID antibody-positive controls. Comparable results of target binding inhibition and similar impacts on exposure suggest that the observed reduction of Cmax and Ctrough levels in patients is caused by the neutralizing potential of ADAs and allows a correlation between ADA response and loss of exposure. Therefore, the described study provides important functional aspects for the development of an appropriately designed PK assay for bispecific antibodies as an alternative option towards understanding the neutralizing ADA impact on exposure.
Collapse
Affiliation(s)
- Gregor P. Lotz
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Achim Lutz
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Meret Martin-Facklam
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Andre Hansbauer
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Eginhard Schick
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Ekkehard Moessner
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Michael Antony
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Thomas Stuchly
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Maria Viert
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Ralf J. Hosse
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | | | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Martin Schäfer
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Mirko Ritter
- Roche Diagnostics GmbH, Antibody Development Technologies, Penzberg, Germany
| | - Kay-Gunnar Stubenrauch
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
19
|
Saha B, Lee JH, Kwon I, Chung H. Site-Specific Conjugation of Bottlebrush Polymers to Therapeutic Protein via Bioorthogonal Chemistry. Biomacromolecules 2024; 25:3200-3211. [PMID: 38591457 DOI: 10.1021/acs.biomac.4c00359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Achieving efficient and site-specific conjugation of therapeutic protein to polymer is crucial to augment their applicability in the realms of biomedicine by improving their stability and enzymatic activity. In this study, we exploited tetrazine bioorthogonal chemistry to achieve the site-specific conjugation of bottlebrush polymers to urate oxidase (UOX), a therapeutic protein for gout treatment. An azido-functionalized zwitterionic bottlebrush polymer (N3-ZBP) using a "grafting-from" strategy involving RAFT and ATRP methods was synthesized, and a trans-cyclooctene (TCO) moiety was introduced at the polymer end through the strain-promoted azide-alkyne click (SPAAC) reaction. The subsequent coupling between TCO-incorporated bottlebrush polymer and tetrazine-labeled UOX using a fast and safe bioorthogonal reaction, inverse electron demand Diels-Alder (IEDDA), led to the formation of UOX-ZBP conjugates with a 52% yield. Importantly, the enzymatic activity of UOX remained unaffected following polymer conjugation, suggesting a minimal change in the folded structure of UOX. Moreover, UOX-ZBP conjugates exhibited enhanced proteolytic resistance and reduced antibody binding, compared to UOX-wild type. Overall, the present findings reveal an efficient and straightforward route for synthesizing protein-bottlebrush polymer conjugates without compromising the enzymatic activity while substantially reducing proteolytic degradation and antibody binding.
Collapse
Affiliation(s)
- Biswajit Saha
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
| | - Jae Hun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Inchan Kwon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Hoyong Chung
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida 32310, United States
| |
Collapse
|
20
|
Asar M, Newton-Northup J, Soendergaard M. Improving Pharmacokinetics of Peptides Using Phage Display. Viruses 2024; 16:570. [PMID: 38675913 PMCID: PMC11055145 DOI: 10.3390/v16040570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Phage display is a versatile method often used in the discovery of peptides that targets disease-related biomarkers. A major advantage of this technology is the ease and cost efficiency of affinity selection, also known as biopanning, to identify novel peptides. While it is relatively straightforward to identify peptides with optimal binding affinity, the pharmacokinetics of the selected peptides often prove to be suboptimal. Therefore, careful consideration of the experimental conditions, including the choice of using in vitro, in situ, or in vivo affinity selections, is essential in generating peptides with high affinity and specificity that also demonstrate desirable pharmacokinetics. Specifically, in vivo biopanning, or the combination of in vitro, in situ, and in vivo affinity selections, has been proven to influence the biodistribution and clearance of peptides and peptide-conjugated nanoparticles. Additionally, the marked difference in properties between peptides and nanoparticles must be considered. While peptide biodistribution depends primarily on physiochemical properties and can be modified by amino acid modifications, the size and shape of nanoparticles also affect both absorption and distribution. Thus, optimization of the desired pharmacokinetic properties should be an important consideration in biopanning strategies to enable the selection of peptides and peptide-conjugated nanoparticles that effectively target biomarkers in vivo.
Collapse
Affiliation(s)
- Mallika Asar
- College of Osteopathic Medicine, Kansas City University, Kansas City, MO 64106, USA;
| | | | - Mette Soendergaard
- Cell Origins LLC, 1601 South Providence Road Columbia, Columbia, MO 65203, USA;
- Department of Chemistry, Western Illinois University, Macomb, IL 61455, USA
| |
Collapse
|
21
|
Cohen JR, Brych SR, Prabhu S, Bi V, Elbaradei A, Tokuda JM, Xiang C, Hokom M, Cui X, Ly C, Amos N, Sun J, Calamba D, Herskovitz J, Capili A, Nourbakhsh K, Merlo A, Carreon J, Wypych J, Narhi LO, Jawa V, Joubert MK. A High Threshold of Biotherapeutic Aggregate Numbers is Needed to Induce an Immunogenic Response In Vitro, In Vivo, and in the Clinic. Pharm Res 2024; 41:651-672. [PMID: 38519817 DOI: 10.1007/s11095-024-03678-2] [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: 09/29/2023] [Accepted: 02/15/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND AND PURPOSE There is concern that subvisible aggregates in biotherapeutic drug products pose a risk to patient safety. We investigated the threshold of biotherapeutic aggregates needed to induce immunogenic responses. METHODS AND RESULTS Highly aggregated samples were tested in cell-based assays and induced cellular responses in a manner that depended on the number of particles. The threshold of immune activation varied by disease state (cancer, rheumatoid arthritis, allergy), concomitant therapies, and particle number. Compared to healthy donors, disease state patients showed an equal or lower response at the late phase (7 days), suggesting they may not have a higher risk of responding to aggregates. Xeno-het mice were used to assess the threshold of immune activation in vivo. Although highly aggregated samples (~ 1,600,000 particles/mL) induced a weak and transient immunogenic response in mice, a 100-fold dilution of this sample (~ 16,000 particles/mL) did not induce immunogenicity. To confirm this result, subvisible particles (up to ~ 18,000 particles/mL, containing aggregates and silicone oil droplets) produced under representative administration practices (created upon infusion of a drug product through an IV catheter) did not induce a response in cell-based assays or appear to increase the rate of adverse events or immunogenicity during phase 3 clinical trials. CONCLUSION The ability of biotherapeutic aggregates to elicit an immune response in vitro, in vivo, and in the clinic depends on high numbers of particles. This suggests that there is a high threshold for aggregates to induce an immunogenic response which is well beyond that seen in standard biotherapeutic drug products.
Collapse
Affiliation(s)
- Joseph R Cohen
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| | - Stephen R Brych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Siddharth Prabhu
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vivian Bi
- The Department of Biosimilars, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Ahmed Elbaradei
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Joshua M Tokuda
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Cathie Xiang
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Martha Hokom
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of BioAnalytical Sciences, Genentech, Inc, South San Francisco, CA, 94080, USA
| | - Xiaohong Cui
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Claudia Ly
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Nathan Amos
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jilin Sun
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Dominador Calamba
- Translational Safety and Bioanalytical Sciences, Amgen Inc, Thousand Oaks, CA, 91320, USA
| | - Jonathan Herskovitz
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Allyson Capili
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Kimya Nourbakhsh
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Anthony Merlo
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Julia Carreon
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Jette Wypych
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Linda O Narhi
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Vibha Jawa
- The Department of Clinical Immunology, Amgen Inc, Thousand Oaks, CA, 91320, USA
- Department of Pharmacometrics, Disposition & Bioanalysis, Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | - Marisa K Joubert
- The Department of Process Development, Amgen Inc, One Amgen Center Dr, Thousand Oaks, CA, 91320, USA.
| |
Collapse
|
22
|
Biesdorf C, Guan X, Siddani SR, Hoffman D, Boehm N, Medeiros BC, Doi T, de Jonge M, Rasco D, Menon RM, Polepally AR. Pharmacokinetics and immunogenicity of eftozanermin alfa in subjects with previously-treated solid tumors or hematologic malignancies: results from a phase 1 first-in-human study. Cancer Chemother Pharmacol 2024; 93:329-339. [PMID: 38036720 DOI: 10.1007/s00280-023-04613-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023]
Abstract
PURPOSE Eftozanermin alfa is a second-generation tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor agonist that enhances death receptor 4/5 clustering on tumor cells to induce apoptosis. We report the pharmacokinetics and immunogenicity of eftozanermin alfa administered intravenously to 153 adults with previously-treated solid tumors or hematologic malignancies from the first-in-human, open-label, dose-escalation and dose-optimization study. METHODS Dose escalation evaluated eftozanermin alfa monotherapy 2.5-15 mg/kg on Day 1 or Days 1/8 of a 21-day cycle. Dose optimization evaluated eftozanermin alfa monotherapy or combination therapy with either oral venetoclax 400-800 mg daily (eftozanermin alfa 1.25-7.5 mg/kg Days 1/8/15 of a 21-day cycle) or chemotherapy (eftozanermin alfa 3.75 or 7.5 mg/kg Days 1/8/15/22 of a 28-day cycle and FOLFIRI regimen [leucovorin, 5-fluorouracil, and irinotecan] with/without bevacizumab on Days 1/15 of a 28-day cycle). RESULTS Systemic exposures (maximum observed concentration [Cmax] and area under the concentration-time curve [AUC]) of eftozanermin alfa were approximately dose-proportional across the entire dose escalation range with minimal to no accumulation in Cycle 3 versus Cycle 1 exposures. Comparable exposures and harmonic mean half-lives (35.1 h [solid tumors], 31.3 h [hematologic malignancies]) were observed between malignancy types. Exposures (dose-normalized Cmax and AUC) in Japanese subjects were similar to non-Japanese subjects. Furthermore, eftozanermin alfa/venetoclax combination therapy did not have an impact on the exposures of either agent. Treatment-emergent anti-drug antibodies were observed in 9.4% (13/138) of subjects. CONCLUSIONS The study results, including a pharmacokinetic profile consistent with weekly dosing and low incidence of immunogenicity, support further investigation of eftozanermin alfa. TRIAL REGISTRATION ID NCT03082209.
Collapse
Affiliation(s)
- Carla Biesdorf
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA.
| | - Xiaowen Guan
- AbbVie Biotherapeutics Inc., South San Francisco, CA, USA
| | - Satya R Siddani
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | - David Hoffman
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | | | | | - Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | | | - Drew Rasco
- South Texas Accelerated Research Therapeutics (START), San Antonio, TX, USA
| | - Rajeev M Menon
- Clinical Pharmacology, AbbVie Inc., 1 North Waukegan Road, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | | |
Collapse
|
23
|
Opolka-Hoffmann E, Edelmann MR, Otteneder MB, Hauri S, Jordan G, Schrag P, Lechmann M, Winter G, Staack RF. Biodistribution of Drug/ADA Complexes: The Impact of Immune Complex Formation on Antibody Distribution. AAPS J 2024; 26:33. [PMID: 38478197 DOI: 10.1208/s12248-024-00899-6] [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: 12/08/2023] [Accepted: 02/10/2024] [Indexed: 04/11/2024] Open
Abstract
The clinical use of therapeutic monoclonal antibodies (mAbs) for the treatment of cancer, inflammation, and other indications has been successfully established. A critical aspect of drug-antibody pharmacokinetics is immunogenicity, which triggers an immune response via an anti-drug antibody (ADA) and forms drug/ADA immune complexes (ICs). As a consequence, there may be a reduced efficacy upon neutralization by ADA or an accelerated drug clearance. It is therefore important to understand immunogenicity in biological therapies. A drug-like immunoglobulin G (IgG) was radiolabeled with tritium, and ICs were formed using polyclonal ADA, directed against the complementary-determining region of the drug-IgG, to investigate in vivo biodistribution in rodents. It was demonstrated that 65% of the radioactive IC dose was excreted within the first 24 h, compared with only 6% in the control group who received non-complexed 3H-drug. Autoradiographic imaging at the early time point indicated a deposition of immune complexes in the liver, lung, and spleen indicated by an increased radioactivity signal. A biodistribution study confirmed the results and revealed further insights regarding excretion and plasma profiles. It is assumed that the immune complexes are readily taken up by the reticuloendothelial system. The ICs are degraded proteolytically, and the released radioactively labeled amino acids are redistributed throughout the body. These are mainly renally excreted as indicated by urine measurements or incorporated into protein synthesis. These biodistribution studies using tritium-labeled immune complexes described in this article underline the importance of understanding the immunogenicity induced by therapeutic proteins and the resulting influence on biological behavior.
Collapse
Affiliation(s)
- Eugenia Opolka-Hoffmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany.
| | - Martin R Edelmann
- Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, UK
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Therapeutic Modalities, CH-4070, Basel, Switzerland
| | - Michael B Otteneder
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070, Basel, Switzerland
| | - Simon Hauri
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070, Basel, Switzerland
| | - Gregor Jordan
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany
| | - Peter Schrag
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070, Basel, Switzerland
| | - Martin Lechmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians University, DE-80539, Munich, Germany
| | - Roland F Staack
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, DE-82377, Penzberg, Germany
| |
Collapse
|
24
|
Nwabufo CK. Mirvetuximab soravtansine in ovarian cancer therapy: expert opinion on pharmacological considerations. Cancer Chemother Pharmacol 2024; 93:89-105. [PMID: 37594572 DOI: 10.1007/s00280-023-04575-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
ImmunoGen developed mirvetuximab soravtansine as an antibody-drug conjugate comprising of a humanized anti-folate receptor-α (FRα) monoclonal antibody of IgG1k subtype, a cleavable linker, and a cytotoxic payload, DM4. Mirvetuximab soravtansine was granted accelerated approval by the US FDA on November 14, 2022, for the treatment of adult patients with FRα positive, platinum-resistant epithelial ovarian, fallopian tube or primary peritoneal cancer who have received 1-3 prior systemic treatment regimens. The approval of mirvetuximab soravtansine represents a breakthrough for addressing the unmet medical needs of ovarian cancer, especially for up to 80% of patients who relapse and become resistant to platinum-based chemotherapy, resulting in poor prognosis and limited treatment options. However, it is my impression that addressing several pharmacological factors could improve the safety and efficacy of mirvetuximab soravtansine. This article summarizes the current pharmacological profile of mirvetuximab soravtansine and provides an expert opinion on pharmacological strategies for optimizing its safety and efficacy profile for the treatment of platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Chukwunonso K Nwabufo
- OneDrug, Toronto, ON, Canada.
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada.
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
25
|
Brekkan A, Lledo-Garcia R, Lacroix B, Jönsson S, Karlsson MO, Plan EL. Characterization of anti-drug antibody dynamics using a bivariate mixed hidden-markov model by nonlinear-mixed effects approach. J Pharmacokinet Pharmacodyn 2024; 51:65-75. [PMID: 37943398 PMCID: PMC10884144 DOI: 10.1007/s10928-023-09890-8] [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: 03/31/2023] [Accepted: 10/01/2023] [Indexed: 11/10/2023]
Abstract
Biological therapies may act as immunogenic triggers leading to the formation of anti-drug antibodies (ADAs). Population pharmacokinetic (PK) models can be used to characterize the relationship between ADA and drug disposition but often rely on the ADA bioassay results, which may not be sufficiently sensitive to inform on this characterization.In this work, a methodology that could help to further elucidate the underlying ADA production and impact on the drug disposition was explored. A mixed hidden-Markov model (MHMM) was developed to characterize the underlying (hidden) formation of ADA against the biologic, using certolizumab pegol (CZP), as a test drug. CZP is a PEGylated Fc free TNF-inhibitor used in the treatment of rheumatoid arthritis and other chronic inflammatory diseases.The bivariate MHMM used information from plasma drug concentrations and ADA measurements, from six clinical studies (n = 845), that were correlated through a bivariate Gaussian function to infer about two hidden states; production and no-production of ADA influencing PK. Estimation of inter-individual variability was not supported in this case. Parameters associated with the observed part of the model were reasonably well estimated while parameters associated with the hidden part were less precise. Individual state sequences obtained using a Viterbi algorithm suggested that the model was able to determine the start of ADA production for each individual, being a more assay-independent methodology than traditional population PK. The model serves as a basis for identification of covariates influencing the ADA formation, and thus has the potential to identify aspects that minimize its impact on PK and/or efficacy.
Collapse
Affiliation(s)
- Ari Brekkan
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, SE-75123, Sweden
| | | | | | - Siv Jönsson
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, SE-75123, Sweden
| | - Mats O Karlsson
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, SE-75123, Sweden
| | - Elodie L Plan
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, SE-75123, Sweden.
| |
Collapse
|
26
|
Arjomandi A, Siradze K, Cheu M, Davancaze T, Yadav R, Rao GK, Wong L, Fischer SK. Successful Development of Nonclinical Anti-Drug Antibody Assays to Support Zinpentraxin Alfa Reproductive Toxicology Studies. AAPS J 2024; 26:16. [PMID: 38267613 DOI: 10.1208/s12248-024-00891-0] [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: 09/22/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Immunogenicity assessment is an essential part of biotherapeutic drug development. While the immune response in animals is not always representative of the human immune response, immunogenicity data obtained in animal models is still informative for the evaluation of drug exposure and safety. The most common assay format used for the detection of anti-drug antibodies (ADAs) in preclinical and clinical studies is the bridging format. The advantage of this method is that it can detect all antibody isotypes generated against the therapeutic. However, the method development can be time-consuming and labor-intensive, due to the need for labeling of the drug which is used both as capture and detection. Various generic ADA assays have been successfully implemented to overcome these disadvantages and to enable faster assay development timelines to support nonclinical toxicology studies. Here, we describe the challenges in the development of an assay to detect antibodies to zinpentraxin alfa, a recombinant human pentraxin-2, in rabbit and rat toxicology studies. Our initial efforts to develop a bridging assay failed, prompting us to develop a method adapted from generic assay formats to detect anti-zinpentraxin alfa antibodies in the serum of different species with minimal optimization. However, while the general assay format remained similar, assay reagents were adapted between the different species, resulting in the development of two distinct assays for the detection of ADAs in rat and rabbit. Here, we share the final development/validation data and the immunogenicity study results. Our work highlights the need for the evaluation of alternate assay formats when evaluating novel drug modalities.
Collapse
Affiliation(s)
- Audrey Arjomandi
- Department of BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Ketevan Siradze
- Department of BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Melissa Cheu
- Department of BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Teresa Davancaze
- Department of BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Rajbharan Yadav
- Department of Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Gautham K Rao
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Lisa Wong
- Department of Safety Assessment, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Saloumeh K Fischer
- Department of BioAnalytical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| |
Collapse
|
27
|
McClellan JE, Ómarsdóttir S, Roy N, Berger V, Michel C, Berti F. The totality of evidence approach in the development of AVT02 (adalimumab), a biosimilar to Humira. Ther Adv Chronic Dis 2024; 15:20406223231223286. [PMID: 38250743 PMCID: PMC10799607 DOI: 10.1177/20406223231223286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
The development of a biosimilar is based on comparative structural, physicochemical, functional and clinical assessments. The sum of these analyses encompasses the 'totality of evidence', which demonstrates no clinically meaningful differences between the biosimilar and the reference product (RP). Once biosimilarity has been established, provided there is suitable scientific justification, clinical data may be extrapolated to other indications of the RP. AVT02 has been developed as a biosimilar to high-concentration, low-volume Humira (adalimumab), an anti-tumour necrosis factor-alpha monoclonal antibody approved for various chronic inflammatory indications. The totality of evidence for AVT02 is described, supporting its approval as an adalimumab biosimilar for all approved indications globally. Analytical similarity assessments using mass spectrometry methods demonstrated identical amino acid sequences for AVT02 and the RP, with high similarity in terms of primary structure, post-translational modifications and higher-order structural attributes. The mechanism of action was assessed by various cell-based potency assays and binding assays, and the results demonstrated that AVT02 is highly similar to the RP. No clinically meaningful differences in terms of purity, potency and safety were observed, and minor differences in a few physiochemical attributes did not impact the in vitro biologic activity and were not considered clinically relevant. Clinical similarity was demonstrated by comparing the pharmacokinetic, efficacy, safety and immunogenicity profiles of AVT02 with those of the RP. Clinical studies supported similar pharmacokinetic and comparable immunogenicity profiles between AVT02 and the RP in healthy participants and participants with moderate-to-severe chronic plaque psoriasis, with no new safety signals detected. The totality of evidence described demonstrates the biosimilarity of AVT02 to the RP, thereby fulfilling the scientific and regulatory requirements for AVT02 as a high-concentration biosimilar for the treatment of chronic plaque psoriasis and all approved indications of the RP.
Collapse
Affiliation(s)
| | - Sesselja Ómarsdóttir
- Alvotech hf., Reykjavík, Iceland
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavík, Iceland
| | | | | | | | | |
Collapse
|
28
|
Sun R, Qian MG, Zhang X. T and B cell epitope analysis for the immunogenicity evaluation and mitigation of antibody-based therapeutics. MAbs 2024; 16:2324836. [PMID: 38512798 PMCID: PMC10962608 DOI: 10.1080/19420862.2024.2324836] [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: 10/06/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
The surge in the clinical use of therapeutic antibodies has reshaped the landscape of pharmaceutical therapy for many diseases, including rare and challenging conditions. However, the administration of exogenous biologics could potentially trigger unwanted immune responses such as generation of anti-drug antibodies (ADAs). Real-world experiences have illuminated the clear correlation between the ADA occurrence and unsatisfactory therapeutic outcomes as well as immune-related adverse events. By retrospectively examining research involving immunogenicity analysis, we noticed the growing emphasis on elucidating the immunogenic epitope profiles of antibody-based therapeutics aiming for mechanistic understanding the immunogenicity generation and, ideally, mitigating the risks. As such, we have comprehensively summarized here the progress in both experimental and computational methodologies for the characterization of T and B cell epitopes of therapeutics. Furthermore, the successful practice of epitope-driven deimmunization of biotherapeutics is exceptionally highlighted in this article.
Collapse
Affiliation(s)
- Ruoxuan Sun
- Global Drug Metabolism, Pharmacokinetics & Modeling, Preclinical & Translational Sciences, Takeda Development Center Americas, Inc. (TDCA), Cambridge, MA, USA
| | - Mark G. Qian
- Global Drug Metabolism, Pharmacokinetics & Modeling, Preclinical & Translational Sciences, Takeda Development Center Americas, Inc. (TDCA), Cambridge, MA, USA
| | - Xiaobin Zhang
- Global Drug Metabolism, Pharmacokinetics & Modeling, Preclinical & Translational Sciences, Takeda Development Center Americas, Inc. (TDCA), Cambridge, MA, USA
| |
Collapse
|
29
|
Guerrieri D, Horvat M, Fan J, Wang J, Lemke L, Richter OV, Poetzl J. Signal-to-noise ratio to assess magnitude, kinetics and impact on pharmacokinetics of the immune response to an adalimumab biosimilar. Bioanalysis 2024; 16:33-48. [PMID: 38031738 DOI: 10.4155/bio-2023-0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Abstract
Background: The antidrug antibody (ADA) signal-to-noise (S/N) ratio was explored as a novel immunogenicity measure to evaluate the immune response of healthy subjects to a single dose of GP2017, an adalimumab biosimilar. Methodology/results: Bioanalytical methods used for the analysis of ADA S/N ratios and ADA titers were validated for sensitivity, precision and drug interference. ADA S/N ratios strongly correlated with ADA titers. Correlations between ADA area under the curve and ADAmax and pharmacokinetics (PK) were stronger for ADA S/N ratio than for ADA titers. Conclusion: ADA S/N ratio allowed for a more sensitive evaluation of the magnitude and kinetics of the immune response, was better correlated with adalimumab PK and was superior to ADA titers in assessing the impact of the immune response on PK.
Collapse
Affiliation(s)
- Davide Guerrieri
- Clinical Development Biopharmaceuticals, Hexal AG (A Sandoz company), D-83607 Holzkirchen, Germany
| | - Matej Horvat
- Biosimilar Technical Development, Sandoz, SI-1526 Ljubljana, Slovenia
| | - Jamie Fan
- Clinical Development Biopharmaceuticals, Sandoz Inc., NJ 08540 Princeton, USA
| | - Jessie Wang
- Clinical Development Biopharmaceuticals, Sandoz Inc., NJ 08540 Princeton, USA
| | - Lena Lemke
- Clinical Development Biopharmaceuticals, Hexal AG (A Sandoz company), D-83607 Holzkirchen, Germany
| | - Oliver von Richter
- Clinical Development Biopharmaceuticals, Hexal AG (A Sandoz company), D-83607 Holzkirchen, Germany
| | - Johann Poetzl
- Clinical Development Biopharmaceuticals, Hexal AG (A Sandoz company), D-83607 Holzkirchen, Germany
| |
Collapse
|
30
|
Wen YF, Ji P, Schrieber SJ, Rathi S, McGuirt D, Liu J, Chen J, Wang YM, Doddapaneni S, Sahajwalla C. Evaluation of Truncated AUC as an Alternative Measure to Assess Pharmacokinetic Comparability in Bridging Biologic-Device Using Prefilled Syringes and Autoinjectors. J Clin Pharmacol 2023; 63:1417-1429. [PMID: 37507728 DOI: 10.1002/jcph.2322] [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: 05/03/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
Pharmacokinetic (PK) comparisons between therapeutic biologics have largely been based on the total area under the concentration-time curve (AUC) and the maximum concentration (Cmax ). For biologics with a long half-life, a PK comparability study may be long in duration and costly to conduct. The goal of this study was to evaluate whether a truncated AUC (tAUC) can be used to assess PK comparability when bridging prefilled syringe (PFS) and autoinjector (AI) treatment options for biologics with a long half-life. Fifteen biologics license applications (BLAs) were included to determine the concordance and geometric percent coefficient of variation (%CV) between tAUCs evaluated on days 7, 14, 21, and 28 and AUC evaluated to infinity (AUC0-inf ). Concordance is established if the tAUCs are comparable with AUC0-inf . Trial simulation was performed to examine the effect of the absorption rate constant (ka ) and sample size on the concordance of tAUCs. The tAUCs evaluated on day 14, 21, and 28 had 100% concordance with AUC0-inf for all 15 BLAs. The concordance of tAUC evaluated at day 7 was 87.5%. Based on the trial simulation, tAUC evaluated to day 28 post-dose can achieve high concordance (≥85%) for biologics exhibiting linear or nonlinear elimination with a ka of ≥0.1/day and with a sample size of 70 subjects per arm. tAUC appears to be a promising alternative PK measure, relative to AUC0-inf , for PK comparability assessments.
Collapse
Affiliation(s)
- Ya-Feng Wen
- Division of Inflammation and Immune Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Ping Ji
- Division of Inflammation and Immune Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Sarah J Schrieber
- Office of Therapeutic Biologics and Biosimilars, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Sneha Rathi
- Division of Inflammation and Immune Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Delaney McGuirt
- Division of Inflammation and Immune Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Jiang Liu
- Division of Pharmacometrics, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Jianmeng Chen
- Division of Inflammation and Immune Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Yow-Ming Wang
- Therapeutic Biologics Program, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Suresh Doddapaneni
- Division of Inflammation and Immune Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Chandrahas Sahajwalla
- Division of Inflammation and Immune Pharmacology, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
31
|
Cheng CA, Jiang AL, Liu YR, Chang LC. Investigation of Immunogenicity Assessment of Biosimilar Monoclonal Antibodies in the United States. Clin Pharmacol Ther 2023; 114:1274-1284. [PMID: 37634125 DOI: 10.1002/cpt.3033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023]
Abstract
Immunogenicity is critical for biologics. However, reference biologics labeling documents do not necessarily mention immunogenicity impact, rendering the development of biosimilars more challenging. We aimed to investigate the comparative assessment of immunogenicity profiles between biosimilars and their respective reference biologics in the review reports of the biosimilar monoclonal antibody applications approved by the Center for Drug Evaluation and Research (CDER), US Food and Drug Administration (FDA) as of March 13, 2022, covering 22 applications approved between April 5, 2016, and December 17, 2021. The maximum differences in anti-drug antibody (ADA) and neutralizing antibody (NAb) incidences between biosimilars and reference products mostly fell within ± 15% (-13.6% to 12%) and ± 20% (-17.4% to 17.1%, except extreme values of -23.4% and 66.7%), respectively. In comparison with antineoplastic agents, more immunosuppressants had ADA-positive (11/11, 100.0% vs. 8/10, 80.0%)/NAb-positive (11/11, 100.0% vs. 3/10, 30.0%) subjects, and the distribution of the aforementioned incidence differences was wider. The investigated biosimilars with available data for analysis demonstrated a high degree of consistency with their reference products in terms of the impact on pharmacokinetic parameters. No increase in immunogenicity was found in available switching studies. Most (16/22, 72.7%) biosimilars were issued post-marketing requirements that were not directly related to immunogenicity concerns. The FDA considered the totality of evidence assessing clinical consequences of immunogenicity differences, if any. Additional information on titers and subgroup analysis may be warranted to elucidate the critical attributes of immunogenicity impact and to aid in forming cost-effective strategies for biosimilar development.
Collapse
Affiliation(s)
- Ching-An Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ai-Lei Jiang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Ru Liu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Lin-Chau Chang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
32
|
Miao G, He Y, Lai K, Zhao Y, He P, Tan G, Wang X. Accelerated blood clearance of PEGylated nanoparticles induced by PEG-based pharmaceutical excipients. J Control Release 2023; 363:12-26. [PMID: 37717659 DOI: 10.1016/j.jconrel.2023.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
PEGylated nanomedicines have been extensively developed and applied to cancer therapy. However, the antitumor efficacy of these nanoparticles is hampered by the accelerated blood clearance (ABC) effect caused by anti-PEG antibodies in vivo. There is still limited understanding about the cause of pre-existing anti-PEG antibodies in the human body. Herein, we discovered that PEG-based pharmaceutical excipients, commonly used in clinical and daily settings, could induce anti-PEG antibodies in vivo and lead to considerable potential clinical impacts on pharmacokinetics and pharmacodynamics of PEGylated nanoparticles. Specifically, we investigated the ability of poloxamer 188 (F68) and poloxamer 407 (F127), the two most frequently used PEG-based pharmaceutical excipients, to elicit the production of anti-PEG antibodies and influence the pharmacokinetics of PEGylated nanoparticles, with PEGylated liposome nanoparticles (L-NPs) as a model. Anti-PEG IgG and IgM levels were significantly boosted 3.8- and 32.2-fold, respectively, after pre-injection with F68, leading to rapid clearance of subsequently injected L-NPs from circulation due to the capture by neutrophils and monocytes. However, pre-injection of F127 did not induce the production of anti-PEG IgG, although there was a 7.7-fold increase in IgM level, which resulted in minimal effect on circulation time of L-NPs. Furthermore, the potential clinical impacts of F68 and F127 were further inspected for PEGylated liposomal doxorubicin (PLD). It was found that administering F68 prior to treatment led to over a one-third decrease in the antitumor effectiveness of PLD, while F127 had a negligible impact. Our study elucidates the mechanism by which PEG-based pharmaceutical excipients influence the effectiveness of PEGylated nanomedicines. It also highlights the significance of considering the potential for an ABC effect induced by PEG-based pharmaceutical excipients in patients.
Collapse
Affiliation(s)
- Guifeng Miao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Yuejian He
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Keren Lai
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Yan Zhao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Peiyi He
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Guozhu Tan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China
| | - Xiaorui Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515 Guangzhou, Guangdong Province, China.
| |
Collapse
|
33
|
Penny HL, Hainline K, Theoharis N, Wu B, Brandl C, Webhofer C, McComb M, Wittemer-Rump S, Koca G, Stienen S, Bargou RC, Hummel HD, Loidl W, Grüllich C, Eggert T, Tran B, Mytych DT. Characterization and root cause analysis of immunogenicity to pasotuxizumab (AMG 212), a prostate-specific membrane antigen-targeting bispecific T-cell engager therapy. Front Immunol 2023; 14:1261070. [PMID: 37942314 PMCID: PMC10628759 DOI: 10.3389/fimmu.2023.1261070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction In oncology, anti-drug antibody (ADA) development that significantly curtails response durability has not historically risen to a level of concern. The relevance and attention ascribed to ADAs in oncology clinical studies have therefore been limited, and the extant literature on this subject scarce. In recent years, T cell engagers have gained preeminence within the prolific field of cancer immunotherapy. These drugs whose mode of action is expected to potently stimulate anti-tumor immunity, may potentially induce ADAs as an unintended corollary due to an overall augmentation of the immune response. ADA formation is therefore emerging as an important determinant in the successful clinical development of such biologics. Methods Here we describe the immunogenicity and its impact observed to pasotuxizumab (AMG 212), a prostate-specific membrane antigen (PSMA)-targeting bispecific T cell engager (BiTE®) molecule in NCT01723475, a first-in-human (FIH), multicenter, dose-escalation study in patients with metastatic castration-resistant prostate cancer (mCRPC). To explain the disparity in ADA incidence observed between the SC and CIV arms of the study, we interrogated other patient and product-specific factors that may have explained the difference beyond the route of administration. Results Treatment-emergent ADAs (TE-ADA) developed in all subjects treated with at least 1 cycle of AMG 212 in the subcutaneous (SC) arm. These ADAs were neutralizing and resulted in profound exposure loss that was associated with contemporaneous reversal of initial Prostate Surface Antigen (PSA) responses, curtailing durability of PSA response in patients. Pivoting from SC to a continuous intravenous (CIV) administration route remarkably yielded no subjects developing ADA to AMG 212. Through a series of stepwise functional assays, our investigation revealed that alongside a more historically immunogenic route of administration, non-tolerant T cell epitopes within the AMG 212 amino acid sequence were likely driving the high-titer, sustained ADA response observed in the SC arm. Discussion These mechanistic insights into the AMG 212 ADA response underscore the importance of performing preclinical immunogenicity risk evaluation as well as advocate for continuous iteration to better our biologics.
Collapse
Affiliation(s)
| | - Kelly Hainline
- Department of Clinical Immunology, Amgen, Thousand Oaks, CA, United States
| | | | - Bin Wu
- Department of Biologics, Amgen, Thousand Oaks, CA, United States
| | - Christian Brandl
- Department of Translational Safety & Bioanalytical Sciences, Amgen Research (Munich) GmbH, Munich, Germany
| | - Christian Webhofer
- Department of Process Development, Amgen Research (Munich) GmbH, Munich, Germany
| | - Mason McComb
- Department of Clinical Pharmacology, Modeling & Simulation, Amgen, Thousand Oaks, CA, United States
| | - Sabine Wittemer-Rump
- Bayer AG, Research and Development Oncology (RED Onc), Pharmaceuticals, Berlin, Germany
| | - Gökben Koca
- Bayer AG, Research and Development Oncology (RED Onc), Pharmaceuticals, Berlin, Germany
| | - Sabine Stienen
- Department of Early Development (Oncology), Amgen Research (Munich) GmbH, Munich, Germany
| | - Ralf C. Bargou
- Translational Oncology/Early Clinical Trial Unit (ECTU), Comprehensive Cancer Center Mainfranken, University Hospital Wurzburg, Wurzburg, Germany
| | - Horst-Dieter Hummel
- Translational Oncology/Early Clinical Trial Unit (ECTU), Comprehensive Cancer Center Mainfranken, University Hospital Wurzburg, Wurzburg, Germany
| | - Wolfgang Loidl
- Department of Urology, Ordensklinikum Linz GmbH, Linz, Austria
| | - Carsten Grüllich
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Medical Center, Heidelberg, Germany
| | - Tobias Eggert
- Department of Early Development (Oncology), Amgen, Thousand Oaks, CA, United States
| | - Ben Tran
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Daniel T. Mytych
- Department of Clinical Immunology, Amgen, Thousand Oaks, CA, United States
| |
Collapse
|
34
|
Bader Y, Suliman R, Harb M, Santos V, Al Qaisi I, Alsaadi T. Effectiveness and Safety of Eptinezumab in Episodic and Chronic Migraine Headache in the UAE: A Retrospective Study. Neurol Ther 2023; 12:1683-1693. [PMID: 37418120 PMCID: PMC10444733 DOI: 10.1007/s40120-023-00521-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
INTRODUCTION Eptinezumab is a humanized IgG1 immunoglobulin monoclonal antibody administered intravenously as a preventative migraine treatment. Previously conducted randomized, double-blind, placebo-controlled trials exhibited significant reductions in monthly migraine frequency among adults experiencing episodic and chronic migraine. The present study seeks to expand upon the current findings and to evaluate eptinezumab's efficacy as a preventative treatment for chronic and episodic migraine patients in the United Arab Emirates. This study is intended to represent the first real-world evidence and will hopefully serve as a valuable complement to the existing literature on the subject. METHODS This was a retrospective exploratory study. The participants included within the study were adult (≥ 18 years) patients diagnosed with either episodic or chronic migraine. Patients were categorized according to their history of previous preventative treatment failure. For the final assessment of treatment efficacy, we included only patients with a minimum of 6 months of clinical follow-up data. Patients were assessed at baseline for their monthly migraine frequency and assessed again at months 3 and 6. The primary objective was to evaluate the efficacy of eptinezumab in reducing migraine frequency among chronic and episodic migraine patients. RESULTS A total of 100 participants were identified, of whom 53 completed the study protocol at month 6. Of the total, 40 (75.47%) were female, 46 (86.79%) were Emirati locals, and 16 (30.19%) were pharmaceutically naïve, having never tried any prior preventative therapy. Additionally, 25 (47.17%) patients met the criteria for chronic migraine (CM), whereas the remaining 28 (52.83%) were diagnosed with episodic migraine (EM). The baseline monthly migraine frequency (MMD) was 12.23 (4.97) days across all participants, 15.56 (3.97) for CM patients, and 9.25 (3.76) for EM patients; by month 6, these frequencies reduced to 3.66 (4.21), 4.76 (5.32), and 2.68 (2.61), respectively. Overall, 58.49% of those enrolled experienced > 75% reduction in MMD frequency by month 6. CONCLUSION Patients enrolled in this trial experienced clinically significant reductions in MMD by month 6. Eptinezumab was well tolerated and with one AE of significance that led to discontinuation from the study.
Collapse
Affiliation(s)
- Yazan Bader
- Department of Neurology, American Center for Psychiatry and Neurology, Abu Dhabi, UAE
| | - Reem Suliman
- Department of Neurology, American Center for Psychiatry and Neurology, Abu Dhabi, UAE.
| | - Mohamad Harb
- Department of Neurology, American Center for Psychiatry and Neurology, Abu Dhabi, UAE
| | - Vanessa Santos
- Department of Neurology, American Center for Psychiatry and Neurology, Abu Dhabi, UAE
| | - Ibrahim Al Qaisi
- Department of Neurology, American Center for Psychiatry and Neurology, Abu Dhabi, UAE
| | - Taoufik Alsaadi
- Department of Neurology, American Center for Psychiatry and Neurology, Abu Dhabi, UAE.
| |
Collapse
|
35
|
Lim EHT, Vlaar APJ, de Bruin S, Rückinger S, Thielert C, Habel M, Guo R, Burnett BP, Dickinson J, Brouwer MC, Riedemann NC, van de Beek D. Pharmacokinetic analysis of vilobelimab, anaphylatoxin C5a and antidrug antibodies in PANAMO: a phase 3 study in critically ill, invasively mechanically ventilated COVID-19 patients. Intensive Care Med Exp 2023; 11:37. [PMID: 37332066 DOI: 10.1186/s40635-023-00520-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Vilobelimab, a complement 5a (C5a)-specific monoclonal antibody, reduced mortality in critically ill COVID-19 patients in a phase 3 multicentre, randomized, double-blind, placebo-controlled study. As part of the study, vilobelimab concentrations and C5a levels as well as antidrug antibodies (ADAs) to vilobelimab were analysed. RESULTS From Oct 1, 2020 to Oct 4, 2021, 368 invasively mechanically ventilated COVID-19 patients were randomized: 177 patients were randomly assigned to receive vilobelimab while 191 patients received placebo. Pharmacokinetic sampling was only performed at sites in Western Europe. Blood samples for vilobelimab measurements were available for 93 of 177 (53%) patients in the vilobelimab group and 99 of 191 (52%) patients in the placebo group. On day 8, after three infusions, mean vilobelimab (trough) concentrations ranged from 21,799.3 to 302,972.1 ng/mL (geometric mean 137,881.3 ng/mL). Blood samples for C5a measurements were available for 94 of 177 (53%) patients in the vilobelimab group and 99 of 191 (52%) patients in the placebo group. At screening, C5a levels were highly elevated and comparable between groups. In the vilobelimab group, median C5a levels were 118.3 ng/mL [IQR 71.2-168.2 ng/mL] and in the placebo group, median C5a levels were 104.6 ng/mL [IQR 77.5-156.6 ng/mL]. By day 8, median C5a levels were reduced by 87% in the vilobelimab group (median 14.5 ng/mL [IQR 9.5-21.0 ng/mL], p < 0.001) versus an 11% increase in the placebo group (median 119.2 ng/mL [IQR 85.9-152.1 ng/mL]). Beyond day 8, though plasma sampling was sparse, C5a levels did not reach screening levels in the vilobelimab group while C5a levels remained elevated in the placebo group. Treatment-emergent ADAs were observed in one patient in the vilobelimab group at hospital discharge on day 40 and in one patient in the placebo group at hospital discharge on day 25. CONCLUSIONS This analysis shows that vilobelimab efficiently inhibits C5a in critically ill COVID-19 patients. There was no evidence of immunogenicity associated with vilobelimab treatment. Trial registration ClinicalTrials.gov, NCT04333420. Registered 3 April 2020, https://clinicaltrials.gov/ct2/show/NCT04333420.
Collapse
Affiliation(s)
- Endry H T Lim
- Department of Intensive Care Medicine, Amsterdam UMC Location University of Amsterdam, AMC Room C3-421, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Department of Neurology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
| | - Alexander P J Vlaar
- Department of Intensive Care Medicine, Amsterdam UMC Location University of Amsterdam, AMC Room C3-421, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Sanne de Bruin
- Department of Intensive Care Medicine, Amsterdam UMC Location University of Amsterdam, AMC Room C3-421, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | | | | | | | - Renfeng Guo
- InflaRx Pharmaceuticals Inc, Ann Arbor, MI, USA
| | | | | | - Matthijs C Brouwer
- Department of Neurology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | | | - Diederik van de Beek
- Department of Neurology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Gusarova VD, Smolov MA, Lyagoskin IV, Degterev MB, Rechetnik EV, Rodionov AV, Pantyushenko MS, Shukurov RR. Characterization of a HIR-Fab-IDS, Novel Iduronate 2-Sulfatase Fusion Protein for the Treatment of Neuropathic Mucopolysaccharidosis Type II (Hunter Syndrome). BioDrugs 2023; 37:375-395. [PMID: 37014547 DOI: 10.1007/s40259-023-00590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Mucopolysaccharidosis type II is a severe lysosomal storage disease caused by deficient activity of the enzyme iduronate-2-sulfatase. The only medicinal product approved by the US Food and Drug Administration for enzyme replacement therapy, recombinant iduronate-2-sulfatase (idursulfase, Elaprase®), is a large molecule that is not able to cross the blood-brain barrier and neutralize progressive damage of the central nervous system caused by the accumulation of glycosaminoglycans. Novel chimeric protein HIR-Fab-IDS is an anti-human insulin receptor Fab fragment fused to recombinant modified iduronate-2-sulfatase. This modification provides a highly selective interaction with the human insulin receptor, which leads to the HIR-Fab-IDS crossing the blood-brain barrier owing to internalization of the hybrid molecule by transcytosis into endothelial cells adjacent to the nervous system by the principle of a 'molecular Trojan horse'. OBJECTIVES In this work, the physicochemical and biological characterization of a blood-brain barrier-penetrating fusion protein, HIR-Fab-IDS, is carried out. HIR-Fab-IDS consists of an anti-human insulin receptor Fab fragment fused to recombinant iduronate-2-sulfatase. METHODS Comprehensive analytical characterization utilizing modern techniques (including surface plasmon resonance and mass spectrometry) was performed using preclinical and clinical batches of HIR-Fab-IDS. Critical quality parameters that determine the therapeutic effect of iduronate-2-sulfatase, as well as IDS enzymatic activity and in vitro cell uptake activity were evaluated in comparison with the marketed IDS product Elaprase® (IDS RP). In vivo efficiency of HIR-Fab-IDS in reversing mucopolysaccharidosis type II pathology in IDS-deficient mice was also investigated. The affinity of the chimeric molecule for the INSR was also determined by both an enzyme-linked immunosorbent assay and surface plasmon resonance. We also compared the distribution of 125I-radiolabeled HIR-Fab-IDS and IDS RP in the tissues and brain of cynomolgus monkeys after intravenous administration. RESULTS The HIR-Fab-IDS primary structure investigation showed no significant post-translational modifications that could affect IDS activity, except for the formylglycine content, which was significantly higher for HIR-Fab-IDS compared with that for IDS RP (~ 76.5 vs ~ 67.7%). Because of this fact, the specific enzyme activity of HIR-Fab-IDS was slightly higher than that of IDS RP (~ 2.73 × 106 U/μmol vs ~ 2.16 × 106 U/μmol). However, differences were found in the glycosylation patterns of the compared IDS products, causing a minor reduced in vitro cellular uptake of HIR-Fab-IDS by mucopolysaccharidosis type II fibroblasts compared with IDS RP (half-maximal effective concentration ~ 26.0 vs ~ 23.0 nM). The efficacy of HIR-Fab-IDS in IDS-deficient mice has demonstrated a statistically significant reduction in the level of glycosaminoglycans in the urine and tissues of the main organs to the level of healthy animals. The HIR-Fab-IDS has revealed high in vitro affinity for human and monkey insulin receptors, and the radioactively labeled product has been shown to penetrate to all parts of the brain and peripheral tissues after intravenous administration to cynomolgus monkeys. CONCLUSIONS These findings indicate that HIR-Fab-IDS, a novel iduronate-2-sulfatase fusion protein, is a promising candidate for the treatment of central nervous system manifestations in neurological mucopolysaccharidosis type II.
Collapse
Affiliation(s)
- Valentina D Gusarova
- Pharmaceutical Analysis Department, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy District, Vladimir Region, 601125, Russia.
| | - Maxim A Smolov
- Pharmaceutical Analysis Department, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy District, Vladimir Region, 601125, Russia
| | - Ivan V Lyagoskin
- Pharmaceutical Analysis Department, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy District, Vladimir Region, 601125, Russia
| | - Maksim B Degterev
- Pharmaceutical Analysis Department, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy District, Vladimir Region, 601125, Russia
| | - Elizaveta V Rechetnik
- Department of Scientific Expertise and Pharmacovigilance, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy district, Vladimir Region, 601125, Russia
| | - Alexander V Rodionov
- Pharmaceutical Analysis Department, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy District, Vladimir Region, 601125, Russia
| | - Marina S Pantyushenko
- Pharmaceutical Analysis Department, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy District, Vladimir Region, 601125, Russia
| | - Rahim R Shukurov
- Pharmaceutical Analysis Department, JSC "GENERIUM", 14 Vladimirskaya Street, Volginskiy, Petushinskiy District, Vladimir Region, 601125, Russia
| |
Collapse
|
37
|
Akagi K, Yagishita S, Ohuchi M, Hayashi Y, Takeyasu Y, Masuda K, Shinno Y, Okuma Y, Yoshida T, Goto Y, Horinouchi H, Yamamoto N, Mukae H, Ohe Y, Hamada A. Impact of ramucirumab pharmacokinetics in combination with docetaxel on the efficacy and survival in patients with advanced non-small cell lung cancer. Lung Cancer 2023; 178:247-253. [PMID: 36913912 DOI: 10.1016/j.lungcan.2023.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/14/2023] [Accepted: 03/05/2023] [Indexed: 03/14/2023]
Abstract
OBJECTIVES Ramucirumab, an anti-vascular endothelial growth factor receptor-2 antibody, has been approved for the treatment of non-small cell lung cancer (NSCLC); however, its pharmacokinetic properties in clinical practice are unknown. We aimed to measure ramucirumab concentrations and conduct a retrospective pharmacokinetic analysis using real-world data. MATERIALS AND METHODS Patients with stage III-IV and recurrent NSCLC who received ramucirumab plus docetaxel were evaluated in this study. After the first administration, the ramucirumab trough concentration (Ctrough) was measured using liquid chromatography-mass spectrometry. Patient characteristics, adverse events, tumor response, and survival time were retrospectively extracted from medical records from August 2, 2016 to July 16, 2021. RESULTS A total of 131 patients were examined to assess serum ramucirumab concentrations. Ctrough ranged from below the lower limit of quantification (BLQ) to 48.8 µg/mL (BLQ ≤ 1st quartile (Q1) ≤ 7.34, 7.34 < 2nd quartile (Q2) ≤ 14.7, 14.7 < 3rd quartile (Q3) ≤ 21.9 and 21.9 < 4th quartile (Q4) ≤ 48.8 µg/mL). The overall response rate was significantly higher in Q2-4 than that in Q1 (p = 0.011). The median progression-free survival was marginally longer, and overall survival was significantly longer in Q2-4 (p = 0.009). The Glasgow prognostic score (GPS) in Q1 was significantly higher than in Q2-4 (p = 0.034) and associated with Ctrough (p = 0.002). CONCLUSION Patients with higher ramucirumab exposure had a high ORR and prolonged survival time, whereas patients with lower ramucirumab exposure were characterized by a high GPS and poor prognosis. Cachexia may reduce the exposure level of ramucirumab in certain patients, reducing the clinical benefits of ramucirumab treatment.
Collapse
Affiliation(s)
- Kazumasa Akagi
- Division of Molecular Pharmacology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Mayu Ohuchi
- Division of Molecular Pharmacology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yoshiharu Hayashi
- Division of Molecular Pharmacology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yuki Takeyasu
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Ken Masuda
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yuki Shinno
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Noboru Yamamoto
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|
38
|
A comprehensive regulatory and industry review of modeling and simulation practices in oncology clinical drug development. J Pharmacokinet Pharmacodyn 2023; 50:147-172. [PMID: 36870005 PMCID: PMC10169901 DOI: 10.1007/s10928-023-09850-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023]
Abstract
Exposure-response (E-R) analyses are an integral component in the development of oncology products. Characterizing the relationship between drug exposure metrics and response allows the sponsor to use modeling and simulation to address both internal and external drug development questions (e.g., optimal dose, frequency of administration, dose adjustments for special populations). This white paper is the output of an industry-government collaboration among scientists with broad experience in E-R modeling as part of regulatory submissions. The goal of this white paper is to provide guidance on what the preferred methods for E-R analysis in oncology clinical drug development are and what metrics of exposure should be considered.
Collapse
|
39
|
Yu X, Li T, Shen Z, Jing H, Xie X, Zhou X, Shen Y, Yang Y. The establishment of B cell-deficient Igh-J KO mouse model by gene editing and efficacy evaluation. Int Immunopharmacol 2023; 116:109704. [PMID: 36689847 DOI: 10.1016/j.intimp.2023.109704] [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: 11/16/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023]
Abstract
Over the last few years, immunotherapy has made significant progress in treating various cancers with therapeutic antibodies. However, therapeutic antibodies have been validated for inducing an unintended immune response in human and animal models, which leads to the emergence of anti-drug antibodies (ADAs) and affects their effectiveness and safety. In preclinical research, ADAs production by B cells may accelerate antibody metabolism and result in missing potential candidate molecules. Thus, it is urgent to develop preclinical models that remove only B cells without affecting the function of T and NK cells. Rearrangement of immunoglobulin heavy chain J gene fragment (Igh-J) is the first link in B cell development, and immunotherapies are currently leaning toward combination treatments with PD-1/PD-L1 antibodies, here we created humanized PD-1, PD-L1 and Igh-J knockout (hPD-1/hPD-L1, Igh-J KO) mice and validated by using the reported high immunogenicity drug M7824 (a protein designed to simultaneously block PD-L1 and TGF-β pathways, poorly anti-tumor efficacy in immunocompetent mice). Phenotypic analysis revealed that human PD-1 and PD-L1 were detectable in hPD-1/hPD-L1, Igh-J KO mice, but not mouse IgM and IgD. Igh-J KO depleted B cells while increased the percentage of other immune cell types. Meanwhile, the humanization of PD-1/PD-L1 and Igh-J KO had neither effect on the overall development, differentiation, or distribution of T cell subtypes, nor on the activation of NK and T cells, indicating that mice can be used for T and NK-related immunotherapies. Furthermore, M7824 treatment of these B cell-deficient mice inhibited tumor growth significantly, with higher M7824 analog concentrations and lower ADA-positive rates. These findings demonstrate that Igh-J KO mice are an effective and stable preclinical model for testing drugs based on T and NK cells with high immunogenicity in vivo.
Collapse
Affiliation(s)
- Xingyan Yu
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Tao Li
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Zhiyuan Shen
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Hongyan Jing
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Xiulong Xie
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Xiaofei Zhou
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Yuelei Shen
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China
| | - Yi Yang
- Department of Postgraduate, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China; Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong 226133, People's Republic of China; Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing 102609, People's Republic of China.
| |
Collapse
|
40
|
Bressler NM, Kim T, Oh I, Russo P, Kim MY, Woo SJ. Immunogenicity With Ranibizumab Biosimilar SB11 (Byooviz) and Reference Product Lucentis and Association With Efficacy, Safety, and Pharmacokinetics: A Post Hoc Analysis of a Phase 3 Randomized Clinical Trial. JAMA Ophthalmol 2023; 141:117-127. [PMID: 36520462 PMCID: PMC9857301 DOI: 10.1001/jamaophthalmol.2022.5403] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Importance SB11 and reference ranibizumab (RBZ) are monoclonal anti-vascular endothelial growth factor (VEGF)-A antibodies approved for the treatment of neovascular age-related macular degeneration (nAMD) and other retinal diseases. The association of ranibizumab immunogenicity and treatment outcomes in patients with nAMD is unclear but relevant regarding concerns about immunogenicity of anti-VEGF biological products. Objective To examine the association of immunogenicity to ranibizumab products (SB11 and RBZ) with efficacy, safety, and pharmacokinetics. Design, Setting, and Participants This was a post hoc analysis of a randomized, double-masked, parallel-group phase 3 equivalence study with participants from 75 centers in 9 countries conducted from March 14, 2018, to December 9, 2019. Included were participants 50 years or older with nAMD and active subfoveal choroidal neovascularization lesions. Interventions Intravitreal injection of SB11 or RBZ, 0.5 mg, every 4 weeks through week 48. Main Outcomes and Measures Serum antidrug antibodies (ADAs) were analyzed during the study period until week 52 to measure immunogenicity. Analyses were performed on immunogenicity (overall ADA positivity) with best-corrected visual acuity (BCVA) and central subfield thickness (CST). Adverse events associated with intraocular inflammation (IOI) and serum ranibizumab levels were compared between overall ADA-positive and ADA-negative participants. Results A total of 705 participants (mean [SD] age, 74.1 [8.5] years; 403 female individuals [57.2%]) were included in the study. The overall incidence of ADA-positivity was 32 of 657 (4.9%) at week 52. The least-squares mean (SE) differences between overall ADA-positive and ADA-negative participants up to week 52 for BCVA and CST, respectively, were 1.6 (2.2) letters (95% CI, -2.7 to 5.8; P = .46) and 3 (13) μm (95% CI, -23 to 29; P = .83). IOI-related events occurred in 1 of 32 overall ADA-positive participants (3.1%) and 4 of 620 overall ADA-negative participants (0.6%). Mean (SD) serum ranibizumab concentrations over time were slightly lower in overall ADA-positive participants compared with those of ADA-negative participants, with a maximum value of 1389.3 (875.4) pg/mL at week 16 vs 1665.4 (1124.0) pg/mL at week 36, respectively. Conclusions and Relevance Results of this post hoc analysis of an equivalence trial suggest that immunogenicity was not associated with efficacy and safety of SB11 and RBZ in participants with nAMD. With a low overall ADA incidence, no clear association was identified between overall ADA positivity and pharmacokinetics. These findings support the biosimilarity of SB11 and RBZ, with no safety concern identified for SB11 vs RBZ associated with immunogenicity. Trial Registration ClinicalTrials.gov Identifier: NCT03150589.
Collapse
Affiliation(s)
- Neil M. Bressler
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland,Editor, JAMA Ophthalmology
| | | | - Inkyung Oh
- Samsung Bioepis, Incheon, Republic of Korea
| | | | | | - Se Joon Woo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
41
|
Development of a bridging ELISA for detection of antibodies against ZV0203 in cynomolgus monkey serum. J Pharmacol Toxicol Methods 2023; 119:107210. [PMID: 36028046 DOI: 10.1016/j.vascn.2022.107210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 01/03/2023]
Abstract
Immunogenicity has been a major concern in the safety evaluation of therapeutic proteins. The assessment of the unwanted immunogenicity of the therapeutic proteins performed in animals prior to clinical trials has been a regulatory requirement. In preclinical studies of therapeutic proteins, cynomolgus monkeys are usually the most relevant animal species. ZV0203, a recombinant humanized anti-human epidermal growth factor receptor 2 monoclonal antibody covalently bound to a cytotoxic drug (Duo-5), possesses a novel format of antibody drug conjugates. In this study, we reported the development, validation, and application of a bridging enzyme-linked immunosorbent assay for the detection of antibodies against ZV0203 in cynomolgus monkey serum. Drug interference at low positive control (18.0 ng/mL) and high positive control (130 ng/mL) of anti-ZV0203 antibodies was not observed when ZV0203 concentration is below 1.74 μg/mL and 1.49 μg/mL, respectively. In addition, no interference was found from mouse IgG1, but interference was observed with human IgG1. No effect of hemolysis was found on the analysis results of the testing samples present in 100% pooled rabbit serum containing 2% (V/V) erythrocyte hemolysates. Besides, spiked anti-ZV0203 antibody in rabbit serum was stable after 5 freeze/thaw cycles. The results showed that the method is suitable for the detection of anti-ZV0203 antibodies in cynomolgus monkey serum. The assay was also successfully applied in the repeated dose study of ZV0203.
Collapse
|
42
|
Utility of in silico prediction of target suppression for antibodies against soluble targets: static versus dynamic models. Eur J Clin Pharmacol 2023; 79:137-147. [PMID: 36416938 DOI: 10.1007/s00228-022-03425-9] [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: 07/15/2022] [Accepted: 11/11/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE Antibodies that bind soluble targets such as cytokines belong to an important class of immunotherapies. Target levels can significantly accumulate after antibody administration due to formation of antibody-target complex, accompanied with suppression in free target which is often difficult to measure. Being a surrogate for pharmacodynamic activity, free target suppression is often predicted using in silico tools. The objective of this work is to illustrate the utility of modelling and to compare static versus dynamic models in the prediction of free target suppression. METHODS Using binding principles, we have derived a static equation to predict free target suppression at steady state (FTSS). This equation operates with five input parameters and accounts for target accumulation over time. Its predictivity was compared to a dynamic model and to other existing metrics in literature via simulations and assumptions were illustrated. RESULTS We demonstrated the utility of in silico tools in prediction of free target suppression using static and dynamic models and clarified the assumptions in key input parameters and their limitations. Predicted values using the FTSS equation correlate very well with those from the dynamic model at level > 20% target suppression, relevant for antagonistic antibodies. CONCLUSION In silico tools are needed to predict target suppression by antibody drugs. Static or dynamic models can be used dependant on the scope, available data and undertaken assumptions. These tools can be used to guide discovery and development of antibodies and has the potential to reduce clinical failure.
Collapse
|
43
|
Rana J, Muñoz MM, Biswas M. Oral tolerance to prevent anti-drug antibody formation in protein replacement therapies. Cell Immunol 2022; 382:104641. [PMID: 36402002 PMCID: PMC9730862 DOI: 10.1016/j.cellimm.2022.104641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
Protein based therapeutics have successfully improved the quality of life for patients of monogenic disorders like hemophilia, Pompe and Fabry disease. However, a significant proportion of patients develop immune responses towards intravenously infused therapeutic protein, which can complicate or neutralize treatment and compromise patient safety. Strategies aimed at circumventing immune responses following therapeutic protein infusion can greatly improve therapeutic efficacy. In recent years, antigen-based oral tolerance induction has shown promising results in the prevention and treatment of autoimmune diseases, food allergies and can prevent anti-drug antibody formation to protein replacement therapies. Oral tolerance exploits regulatory mechanisms that are initiated in the gut associated lymphoid tissue (GALT) to promote active suppression of orally ingested antigen. In this review, we outline general perceptions and current knowledge about the mechanisms of oral tolerance, including tissue specific sites of tolerance induction and the cells involved, with emphasis on antigen presenting cells and regulatory T cells. We define several factors, such as cytokines and metabolites that impact the stability and expansion potential of these immune modulatory cells. We highlight preclinical studies that have been performed to induce oral tolerance to therapeutic proteins or enzymes for single gene disorders, such as hemophilia or Pompe disease. These studies mainly utilize a transgenic plant-based system for oral delivery of antigen in conjugation with fusion protein technology that favors the prevention of antigen degradation in the stomach while enhancing uptake in the small intestine by antigen presenting cells and regulatory T cell induction, thereby promoting antigen specific systemic tolerance.
Collapse
Affiliation(s)
- Jyoti Rana
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maite Melero Muñoz
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
44
|
Joyce A, Shea C, You Z, Gorovits B, Lepsy C. Determination of Anti-drug Antibody Affinity in Clinical Study Samples Provides a Tool for Evaluation of Immune Response Maturation. AAPS J 2022; 24:114. [PMID: 36324032 PMCID: PMC9629885 DOI: 10.1208/s12248-022-00759-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Characterization of clinical anti-drug antibody (ADA) responses to biotherapeutics can be important to understanding the consequences of immunogenicity. ADA are expected to be polyclonal, with composition and affinities that evolve over time. Measuring ADA binding affinity can be complicated by the polyclonal nature of response, residual drug in sample, and low ADA levels. We developed a novel workflow to determine the apparent ADA affinity (KD) against a monoclonal antibody biotherapeutic, PF-06480605. An affinity capture elution pre-treatment step was used to isolate ADA and remove residual drug interference from samples. Solution-phase equilibrium incubation was performed using drug and sample ADA as variable and fixed binding interactants, respectively. Unbound ADA concentration was measured using a Singulex Erenna ligand-binding assay (LBA) method. Apparent ADA KD values were calculated using a custom R Shiny algorithm. KD values determined for ADA positive samples showed good correlation with other immunogenicity parameters, including titers and neutralizing antibody (NAb) activity with a general increase in affinity over time, indicative of a maturing immune response. Time of onset of high affinity responses (KD < 100 pM) varied between patients, ranging from 16 to 24 weeks. Antibody responses appeared monophasic at earlier time points, trending towards a biphasic response with a variable transition time and general increase in proportion of high affinity ADA over time. Herein, we provide a novel, sensitive bioanalytical method to determine the KD of ADA in clinical samples. The observed decrease in ADA KD is consistent with evidence of a maturing immune response.
Collapse
Affiliation(s)
- Alison Joyce
- grid.410513.20000 0000 8800 7493Pfizer, Inc., Worldwide Research & Development, Biomedicine Design, 1 Burtt Road, Andover, Massachusetts USA
| | - Christopher Shea
- grid.410513.20000 0000 8800 7493Pfizer, Inc., Worldwide Research & Development, Biomedicine Design, 1 Burtt Road, Andover, Massachusetts USA
| | - Zhiping You
- grid.410513.20000 0000 8800 7493Pfizer, Inc, Worldwide Research & Development, Early Clinical Development, 1 Burtt Road, Andover, Massachusetts USA
| | - Boris Gorovits
- grid.410513.20000 0000 8800 7493Pfizer, Inc., Worldwide Research & Development, Biomedicine Design, 1 Burtt Road, Andover, Massachusetts USA ,grid.510014.1Present Address: Development Sciences, Sana Biotechnology, Inc., 300 Technology Square, Cambridge, Massachusetts USA
| | - Christopher Lepsy
- grid.410513.20000 0000 8800 7493Pfizer, Inc., Worldwide Research & Development, Biomedicine Design, 1 Burtt Road, Andover, Massachusetts USA
| |
Collapse
|
45
|
Dent M, Mayer KL, Verjan Garcia N, Guo H, Kajiura H, Fujiyama K, Matoba N. Impact of glycoengineering and antidrug antibodies on the anticancer activity of a plant-made lectin-Fc fusion protein. PLANT BIOTECHNOLOGY JOURNAL 2022; 20:2217-2230. [PMID: 35900183 PMCID: PMC9616523 DOI: 10.1111/pbi.13902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 06/27/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Plants are an efficient production platform for manufacturing glycoengineered monoclonal antibodies and antibody-like molecules. Avaren-Fc (AvFc) is a lectin-Fc fusion protein or lectibody produced in Nicotiana benthamiana, which selectively recognizes cancer-associated high-mannose glycans. In this study, we report the generation of a glycovariant of AvFc that is devoid of plant glycans, including the core α1,3-fucose and β1,2-xylose residues. The successful removal of these glycans was confirmed by glycan analysis using HPLC. This variant, AvFcΔXF , has significantly higher affinity for Fc gamma receptors and induces higher levels of luciferase expression in an antibody-dependent cell-mediated cytotoxicity (ADCC) reporter assay against B16F10 murine melanoma cells without inducing apoptosis or inhibiting proliferation. In the B16F10 flank tumour mouse model, we found that systemic administration of AvFcΔXF , but not an aglycosylated AvFc variant lacking affinity for Fc receptors, significantly delayed the growth of tumours, suggesting that Fc-mediated effector functions were integral. AvFcΔXF treatment also significantly reduced lung metastasis of B16F10 upon intravenous challenge whereas a sugar-binding-deficient mutant failed to show efficacy. Lastly, we determined the impact of antidrug antibodies (ADAs) on drug activity in vivo by pretreating animals with AvFcΔXF before implanting tumours. Despite a significant ADA response induced by the pretreatment, we found that the activity of AvFcΔXF was unaffected by the presence of these antibodies. These results demonstrate that glycoengineering is a powerful strategy to enhance AvFc's antitumor activity.
Collapse
Affiliation(s)
- Matthew Dent
- Department of Pharmacology and ToxicologyUniversity of Louisville School of MedicineLouisvilleKYUSA
| | - Katarina L. Mayer
- UofL Health – Brown Cancer CenterUniversity of Louisville School of MedicineLouisvilleKYUSA
| | - Noel Verjan Garcia
- UofL Health – Brown Cancer CenterUniversity of Louisville School of MedicineLouisvilleKYUSA
| | - Haixun Guo
- Department of RadiologyUniversity of Louisville School of MedicineLouisvilleKYUSA
- Center for Predictive MedicineUniversity of Louisville School of MedicineLouisvilleKYUSA
| | - Hiroyuki Kajiura
- International Center for BiotechnologyOsaka UniversityOsakaJapan
| | | | - Nobuyuki Matoba
- Department of Pharmacology and ToxicologyUniversity of Louisville School of MedicineLouisvilleKYUSA
- UofL Health – Brown Cancer CenterUniversity of Louisville School of MedicineLouisvilleKYUSA
- Center for Predictive MedicineUniversity of Louisville School of MedicineLouisvilleKYUSA
| |
Collapse
|
46
|
Qin Q, Gong L. Current Analytical Strategies for Antibody-Drug Conjugates in Biomatrices. Molecules 2022; 27:6299. [PMID: 36234836 PMCID: PMC9572530 DOI: 10.3390/molecules27196299] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/27/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a new class of biotherapeutics, consisting of a cytotoxic payload covalently bound to an antibody by a linker. Ligand-binding assay (LBA) and liquid chromatography-mass spectrometry (LC-MS) are the favored techniques for the analysis of ADCs in biomatrices. The goal of our review is to provide current strategies related to a series of bioanalytical assays for pharmacokinetics (PK) and anti-drug antibody (ADA) assessments. Furthermore, the strengths and limitations of LBA and LC-MS platforms are compared. Finally, potential factors that affect the performance of the developed assays are also provided. It is hoped that the review can provide valuable insights to bioanalytical scientists on the use of an integrated analytical strategy involving LBA and LC-MS for the bioanalysis of ADCs and related immunogenicity evaluation.
Collapse
Affiliation(s)
- Qiuping Qin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Likun Gong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Immunoassay and Immunochemistry, Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 101408, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| |
Collapse
|
47
|
Evaluating Antibody Pharmacokinetics as Prerequisite for Determining True Efficacy as Shown by Dual Targeting of PD-1 and CD96. Biomedicines 2022; 10:biomedicines10092146. [PMID: 36140247 PMCID: PMC9495994 DOI: 10.3390/biomedicines10092146] [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: 06/10/2022] [Revised: 07/04/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
One important prerequisite for developing a therapeutic monoclonal antibody is to evaluate its in vivo efficacy. We tested the therapeutic potential of an anti-CD96 antibody alone or in combination with an anti-PD-1 antibody in a mouse colon cancer model. Early anti-PD-1 treatment significantly decreased tumor growth and the combination with anti-CD96 further increased the therapeutic benefit, while anti-CD96 treatment alone had no effect. In late therapeutic settings, the treatment combination resulted in enhanced CD8+ T cell infiltration of tumors and an increased CD8/Treg ratio. Measured anti-PD-1 concentrations were as expected in animals treated with anti-PD-1 alone, but lower at later time points in animals receiving combination treatment. Moreover, anti-CD96 concentrations dropped dramatically after 10 days and were undetectable thereafter in most animals due to the occurrence of anti-drug antibodies that were increasing antibody clearance. Comparison of the anti-PD-1 concentrations with tumor growth showed that higher antibody concentrations in plasma correlated with better therapeutic efficacy. The therapeutic effect of anti-CD96 treatment could not be evaluated, because plasma concentrations were too low. Our findings strongly support the notion of measuring both plasma concentration and anti-drug antibody formation throughout in vivo studies, in order to interpret pharmacodynamic data correctly.
Collapse
|
48
|
Zhou Y, Zhang F, Starcevic Manning M, Hu Z, Hsu CP, Chen PW, Peng C, Loop B, Mytych DT, Paiva da Silva Lima G. Immunogenicity of erenumab: A pooled analysis of six placebo-controlled trials with long-term extensions. Cephalalgia 2022; 42:749-760. [PMID: 35272533 DOI: 10.1177/03331024221075621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Immunogenicity of erenumab, a human anti-calcitonin gene-related peptide receptor monoclonal antibody developed for migraine prevention, has been evaluated throughout clinical development. METHODS Integrated post hoc analysis assessing immunogenicity of erenumab across six clinical trials in patients with episodic and chronic migraine (N = 2985). Anti-erenumab antibody incidence and potential impact on pharmacokinetics, efficacy, and safety were evaluated in short-term (double-blind treatment phase 12-24 weeks) and long-term (double-blind treatment phase plus extensions of up to 5 years) analyses. RESULTS Anti-erenumab binding antibody incidence was low with few patients developing neutralizing antibodies. Antibody responses were mostly transient with low magnitude. Binding antibodies developed as early as 2-4 weeks after the first dose; the majority developed within the first 6 months and very few after the first year. Serum concentrations of erenumab in antibody-positive patients were generally lower than, but within the range of, antibody-negative patients. There was no impact of anti-erenumab antibodies on erenumab efficacy or safety with no differences between antibody-positive and antibody-negative patients in change in monthly migraine days or adverse event rates. CONCLUSIONS This pooled analysis showed that immunogenicity had no meaningful clinical impact on efficacy or safety of erenumab in patients with migraine.Clinical Trial Registration: ClinicalTrials.gov, NCT01952574; ClinicalTrials.gov, NCT02456740; Clinicaltrials.gov NCT02483585; Clinicaltrials.gov, NCT02174861; Clinicaltrials.gov, NCT02630459; Clinicaltrials.gov, NCT03812224.
Collapse
Affiliation(s)
- Yanchen Zhou
- Clinical Immunology, Amgen Inc., South San Francisco, CA, USA
| | - Feng Zhang
- Global Biostatistical Sciences, Amgen, Inc. Thousand Oaks, CA, USA
| | | | - Zheng Hu
- Translational Safety and Bioanalytical Sciences, Amgen, Inc. Thousand Oaks, CA, USA
| | - Cheng-Pang Hsu
- Clinical Pharmacology, Amgen, Inc. Thousand Oaks, CA, USA
| | - Po-Wei Chen
- Clinical Pharmacology, Amgen, Inc. Thousand Oaks, CA, USA
| | - Cheng Peng
- Global Biostatistical Sciences, Amgen, Inc. Thousand Oaks, CA, USA
| | - Brett Loop
- Global Safety, Amgen Inc., Thousand Oaks, CA, USA
| | | | | |
Collapse
|
49
|
Pre-Clinical In-Vitro Studies on Parameters Governing Immune Complex Formation. Pharmaceutics 2022; 14:pharmaceutics14061254. [PMID: 35745826 PMCID: PMC9227392 DOI: 10.3390/pharmaceutics14061254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/06/2023] Open
Abstract
The success of biotherapeutics is often challenged by the undesirable events of immunogenicity in patients, characterized by the formation of anti-drug antibodies (ADA). Under specific conditions, the ADAs recognizing the biotherapeutic can trigger the formation of immune complexes (ICs), followed by cascades of subsequent effects on various cell types. Hereby, the connection between the characteristics of ICs and their downstream impact is still not well understood. Factors governing the formation of ICs and the characteristics of these IC species were assessed systematically in vitro. Classic analytical methodologies such as SEC-MALS and SV-AUC, and the state-of-the-art technology mass photometry were applied for the characterization. The study demonstrates a clear interplay between (1) the absolute concentration of the involved components, (2) their molar ratios, (3) structural features of the biologic, (4) and of its endogenous target. This surrogate study design and the associated analytical tool-box is readily applicable to most biotherapeutics and provides valuable insights into mechanisms of IC formation prior to FIH studies. The applicability is versatile—from the detection of candidates with immunogenicity risks during developability assessment to evaluation of the impact of degraded or post-translationally modified biotherapeutics on the formation of ICs.
Collapse
|
50
|
Gill KL, Jones HM. Opportunities and Challenges for PBPK Model of mAbs in Paediatrics and Pregnancy. AAPS J 2022; 24:72. [PMID: 35650328 DOI: 10.1208/s12248-022-00722-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/20/2022] [Indexed: 12/20/2022] Open
Abstract
New drugs may in some cases need to be tested in paediatric and pregnant patients. However, it is difficult to recruit such patients and there are many ethical issues around their inclusion in clinical trials. Modelling and simulation can help to plan well-designed clinical trials with a reduced number of participants and to bridge gaps where recruitment is difficult. Physiologically based pharmacokinetic (PBPK) models for small molecule drugs have been used to aid study design and dose adjustments in paediatrics and pregnancy, with several publications in the literature. However, published PBPK models for monoclonal antibodies (mAb) in these populations are scarce. Here, the current status of mAb PBPK models in paediatrics and pregnancy is discussed. Seven mAb PBPK models published for paediatrics were found, which report good prediction accuracy across a wide age range. No mAb PBPK models for pregnant women have been published to date. Current challenges to the development of such PBPK models are discussed, including gaps in our knowledge of relevant physiological processes and availability of clinical data to verify models. As the availability of such data increases, it will help to improve our confidence in the PBPK model predictive ability. Advantages for using PBPK models to predict mAb PK in paediatrics and pregnancy are discussed. For example, the ability to incorporate ontogeny and gestational changes in physiology, prediction of maternal, placental and foetal exposure and the ability to make predictions from in vitro and preclinical data prior to clinical data being available.
Collapse
Affiliation(s)
- Katherine L Gill
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK.
| | - Hannah M Jones
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK
| |
Collapse
|