1
|
Grosskopf AK, Ginart AA, Spinosa P, Shivva V. Pharmacokinetics-Based Design of Subcutaneous Controlled Release Systems for Biologics. CPT Pharmacometrics Syst Pharmacol 2025; 14:668-680. [PMID: 39856532 PMCID: PMC12001277 DOI: 10.1002/psp4.13303] [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: 08/02/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
Protein therapeutics have emerged as an exceedingly promising treatment modality in recent times but are predominantly given as intravenous administration. Transitioning to subcutaneous (SC) administration of these therapies could significantly enhance patient convenience by enabling at-home administration, thereby potentially reducing the overall cost of treatment. Approaches that enable sustained delivery of subcutaneously administered biologics offer further advantages in terms of less frequent dosing and better patient compliance. Controlled release technologies, such as hydrogels and subcutaneous implantable technologies, present exciting solutions by enabling the gradual release of biologics from the delivery system. Despite their substantial potential, significant hurdles remain in appropriately applying and integrating these technologies with the ongoing development of complex biologic-based therapies. We evaluate the potential impact of subcutaneously delivered controlled release systems on the downstream pharmacokinetics (PK) of several FDA-approved biologics by employing rigorous mathematical analysis and predictive PK simulations. By leveraging linear time-invariant (LTI) systems theory, we provide a robust framework for understanding and optimizing the release dynamics of these technologies. We demonstrate simple quantitative metrics and approaches that can inform the design and implementation of controlled release technologies. The findings highlight key opportunity areas to reduce dosing frequency, stabilize concentration profiles, and synergize the codelivery of biologics, calling for collaboration between drug delivery and PK scientists to create the most convenient, optimized, and effective precision therapies.
Collapse
Affiliation(s)
- Abigail K. Grosskopf
- Department of Translational Pharmacokinetics and PharmacodynamicsGenentech Inc.South San FranciscoCaliforniaUSA
| | - Antonio A. Ginart
- Department of Electrical EngineeringStanford UniversityStanfordCaliforniaUSA
| | - Phillip Spinosa
- Department of Translational Pharmacokinetics and PharmacodynamicsGenentech Inc.South San FranciscoCaliforniaUSA
| | - Vittal Shivva
- Department of Translational Pharmacokinetics and PharmacodynamicsGenentech Inc.South San FranciscoCaliforniaUSA
| |
Collapse
|
2
|
De Bartolo A, Romeo N, Marrone A, Rago V, Granieri MC, Vommaro ML, Cupelli A, Cerra MC, Indiveri C, Ronca R, Cantile M, Sanna R, Rocca C, Angelone T. A recombinant fragment antigen-binding (Fab) of trastuzumab displays low cytotoxic profile in adult human cardiomyocytes: first evidence and the key implication of FcγRIIA receptor. Acta Pharmacol Sin 2025; 46:618-631. [PMID: 39414958 PMCID: PMC11845480 DOI: 10.1038/s41401-024-01397-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024]
Abstract
Fragment crystallizable gamma receptors (FcγRs) mediate various cellular responses with significant cardiovascular implications. They contribute to the anticancer activity of trastuzumab (TRZ), a recombinant humanized monoclonal antibody that interferes with human epidermal growth factor receptor 2 (HER2), thereby blocking its physiological function in cardiac cells. This is responsible for cardiac complications that hamper TRZ clinical application. In this study we investigated the involvement of FcγRs in the TRZ cardiotoxicity. We used a recombinant antigen-binding fragment (Fab) of TRZ (rFab-HER2) to examine whether the absence of the Fc region resulted in fewer cardiomyocyte toxicity while preserving TRZ's ability to inhibit HER2. When exposed to rFab-HER2, AC16 human adult ventricular cardiomyocytes were less vulnerable to damage and death, than to TRZ. Specifically, TRZ exhibited cytotoxicity at a lower concentration (150 µg/mL, corresponding to ~1 µM) compared to rFab-HER2 (250 µg/mL, corresponding to ~5 µM). Like TRZ, rFab-HER2 negatively modulated HER2 levels in cardiomyocyte (without inducing cytotoxic activity in BJ human fibroblast cells that either did not express or express very low levels of HER2) and inhibited the downstream ERK/AKT cascades. But rFab-HER2 did not alter cardiomyocyte mitochondrial dynamic balance, and affect apoptosis and inflammation, while it limited cytosolic and mitochondrial ROS indicators. On contrary, the Fc region (50-250 μg/mL) exerted direct cytotoxic action on cardiomyocytes (but not on human fibroblasts that lacked Fc receptors). TRZ (150 μg/mL) markedly upregulated the expression level of FcγRIIA (a FcγRs strongly involved in TRZ-induced antibody-dependent cellular toxicity) in cardiomyocytes, whereas the Fab fragment (150 μg/mL) had no effect. Our results demonstrate that Fc region plays an important pathogenic role in TRZ-induced cardiomyocyte toxicity. In addition, targeting FcγRIIA might contribute to the off-target effects of TRZ therapy.
Collapse
Affiliation(s)
- Anna De Bartolo
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Naomi Romeo
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Alessandro Marrone
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Cosenza, Italy
| | - Maria Concetta Granieri
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Maria Luigia Vommaro
- Department of Biology, E. and E. S. (DiBEST), University of Calabria, Rende, Italy
| | - Arianna Cupelli
- Department of Biology, E. and E. S. (DiBEST), Organ and System Physiology Laboratory, University of Calabria, Cosenza, Italy
| | - Maria Carmela Cerra
- Department of Biology, E. and E. S. (DiBEST), Organ and System Physiology Laboratory, University of Calabria, Cosenza, Italy
| | - Cesare Indiveri
- Department of Biology, E. and E. S. (DiBEST), Unit of Biochemistry, Molecular Biotechnology, and Molecular Biology, University of Calabria, Cosenza, Italy
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| | | | | | | | - Carmine Rocca
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy.
- National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Tommaso Angelone
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy.
- National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
3
|
Cunha DR, Segundo MA, Quinaz MB. Electrochemical methods for evaluation of therapeutic monoclonal antibodies: A review. Biosens Bioelectron 2025; 271:116988. [PMID: 39642528 DOI: 10.1016/j.bios.2024.116988] [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: 08/13/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Biopharmaceuticals are complex pharmaceutical drug products produced by biotechnology in living systems. Small changes in the production process can induce differences in the structure of the active ingredient, which may have a strong impact on its pharmacological properties. Therefore, quality assurance of biopharmaceuticals results in a high analytical effort. Strict quality and stability monitoring of potentially critical quality attributes (CQAs) is required. Electrochemical methods have been contributing to the expansion of sensors and biosensors due to their advantages, such as cost-effectiveness and easy operation. Here, we discuss the recent developments in sensors and biosensors using electrochemical techniques employed for the determination of biopharmaceuticals, namely monoclonal antibodies (mAb) and fragments of mAbs. In the frame of this information, this review aims to critically address electrochemical sensors and biosensors for the analysis of biopharmaceuticals reported since 2016. Electrochemical bio(sensors) development has been mainly based on gold and aptamers, respectively, as the most used electrode material and biorecognition element. Also, Bevacizumab (BEVA) was the main therapeutic mAb detected and 69% of the works described a (bio)sensor) that can be applied to therapeutic drug monitoring.
Collapse
Affiliation(s)
- Diana R Cunha
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| | - Marcela A Segundo
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| | - M Beatriz Quinaz
- LAQV, REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
4
|
Berkhout J, Fairman D, van Noort M, van Steeg TJ. A model-based approach using GSK3772847, an anti-interleukin-33 receptor monoclonal antibody, as a showcase to predict SC administration PK and free target dynamics based on PK and total target measurements after IV administration. CPT Pharmacometrics Syst Pharmacol 2025; 14:17-27. [PMID: 39258338 PMCID: PMC11706423 DOI: 10.1002/psp4.13234] [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: 05/16/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
Integrated modeling of the pharmacokinetic (PK) and target binding, by means of a TMDD model, can provide valuable insights into the expected pharmacodynamic (PD) effects of monoclonal antibodies (mAbs). Optimal characterization of the human PK and target binding for mAbs requires data obtained after intravenous (IV) administration which can be combined with subcutaneous (SC) data to further this characterization. Integration of free and/or total target measurements in a population TMDD model will allow quantification of target engagement which is the first step in the cascade leading to efficacy. However, the assays for determination of free target concentrations are analytically challenging and are inherently biased to overpredict the true concentrations in the presence of mAb:target complexes. For that reason, the objective of the current research was to evaluate the predictive value of free target concentrations in a TMDD model developed using PK and total target observations only. Further, a secondary objective was to demonstrate that prediction of SC data is feasible, based on an existing IV model and typical values of mAb parameters reported for SC absorption. GSK3772847, a human immunoglobulin G2 sigma isotype (IgG2f) mAb that binds to the extracellular domain of the interleukin-33 receptor (IL-33R or ST2) and neutralizes IL-33-mediated ST2 signaling, was used as a model compound for mAbs in this study.
Collapse
Affiliation(s)
- Jan Berkhout
- Leiden Experts on Advanced Pharmacokinetics and Pharmacodynamics (LAP&P)LeidenThe Netherlands
| | - Dave Fairman
- Clinical Pharmacology Modelling and SimulationGSKStevenageHertfordshireUK
| | - Martijn van Noort
- Leiden Experts on Advanced Pharmacokinetics and Pharmacodynamics (LAP&P)LeidenThe Netherlands
| | - Tamara J. van Steeg
- Leiden Experts on Advanced Pharmacokinetics and Pharmacodynamics (LAP&P)LeidenThe Netherlands
| |
Collapse
|
5
|
Luo X, Wang N, Xing Y, Gao X, Yu Y, Liu T, Jiang S, Dong M. Pharmacokinetics of trastuzumab and its efficacy and safety in HER2-positive cancer patients. Cancer Chemother Pharmacol 2024; 94:721-732. [PMID: 39177768 DOI: 10.1007/s00280-024-04707-y] [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: 01/15/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024]
Abstract
Trastuzumab is a potent targeted therapy drug for HER2-positive cancer patients. A comprehensive understanding of trastuzumab's mechanism of action, pharmacokinetic (PK) parameters, and steady-state exposure in different treatment regimens and administration routes is essential for a thorough evaluation of the drug's safety and effectiveness. Due to the distinctive pharmacokinetics, indications, and administration methods of trastuzumab, this understanding becomes crucial. Drug exposure can be assessed by measuring trastuzumab's peak concentration, trough concentration, or area under the curve through assays like enzyme-linked immunosorbent assay (ELISA) or liquid chromatography-tandem mass spectrometry (LC-MS/MS). The dose-response (D-R) and exposure-response (E-R) relationships establish the correlation between drug dosage/exposure and the therapeutic effect and safety. Additionally, various covariates such as body weight, aspartate transaminase, and albumin levels can influence drug exposure. This review provides a comprehensive overview of trastuzumab's mechanism of action, data on steady-state concentration and PK parameters under multiple administration routes and indications, discussions on factors influencing PK parameters, and evaluations of the effectiveness and safety of E-R and D-R in diverse HER2-positive cancer patients.
Collapse
Affiliation(s)
- Xinyu Luo
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Nan Wang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yue Xing
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Xinyue Gao
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yang Yu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Tong Liu
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Shuai Jiang
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Mei Dong
- Department of Pharmacy, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
6
|
Otoya I, Valdiviezo N, Morante Z, Calle C, Ferreyra Y, Huarcaya-Chombo N, Polo-Mendoza G, Castañeda C, Vidaurre T, Neciosup SP, Calderón MJ, Gomez HL. Subcutaneous Trastuzumab: An Observational Study of Safety and Tolerability in Patients With Early HER2-Positive Breast Cancer. Int J Breast Cancer 2024; 2024:9551710. [PMID: 38962673 PMCID: PMC11222001 DOI: 10.1155/2024/9551710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Purpose: In Peru, breast cancer (BC) stands as the most predominant malignancy neoplasm among women. Trastuzumab has marked a significant milestone in the management of this disease. It has been shown to improve prognosis in human epidermal growth factor receptor 2 (HER2)-expressing female patients, but its repercussions and efficacy are yet to be analyzed in a context with limited resources. Methods: The study population is made of woman patients aged 18 years and older diagnosed with HER2-positive BC at Instituto Nacional de Enfermedades Neoplásicas (INEN, Lima, Peru) during 2019-2021 and treated with at least one dose of subcutaneous trastuzumab. We reviewed medical records to register treatment characteristics, adverse events (AEs), disease progression, and survival status. We considered a median follow-up time of 36 and 45 months for progression and survival status. Results: The majority of patients were over 50 years old (54.29%). Tumor size averaged 19.7 ± 16.1 mm. Lymph nodes were present in 44.78% of patients. Most patients received adjuvant chemotherapy (63.8%) as first-line treatment. Descriptive analyses of treatment outcomes revealed a 30% toxicity rate, primarily attributed to arthralgia (47.62%), followed by diarrhea, fatigue, and injection site reactions, with relatively lower discontinuation rates compared to larger scale studies. Differences in demographic, clinical, and treatment characteristics were not statistically significant concerning the emergence of AEs (p > 0.05). Progression appeared in nine patients, and the overall survival (OS) rate stood at 98.6% and 92.8%, respectively, during a median follow-up of 36 and 45 months. Conclusion: The research suggests that subcutaneous trastuzumab is comparable in effectiveness and safety to the intravenous administration. Regional-specific studies may provide valuable insights into demographic factors influencing treatment outcomes in Peru or other countries. Furthermore, it could represent a more accessible alternative, potentially enhancing patient adherence and optimizing healthcare resource logistics.
Collapse
Affiliation(s)
- Iris Otoya
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Natalia Valdiviezo
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Zaida Morante
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Cindy Calle
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Yomali Ferreyra
- Department of BioengineeringUniversidad de Ingeniería y Tecnología, Lima, Peru
| | | | | | - Carlos Castañeda
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Tatiana Vidaurre
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Silvia P. Neciosup
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
| | - Mónica J. Calderón
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
- Institute of Investigations in Biomedical Sciences (INICIB)Universidad Ricardo Palma, Lima, Peru
| | - Henry L. Gomez
- Department of Medical OncologyInstituto Nacional de Enfermedades Neoplásicas, Lima, Peru
- Institute of Investigations in Biomedical Sciences (INICIB)Universidad Ricardo Palma, Lima, Peru
- OncosaludAuna Ideas, Lima, Peru
| |
Collapse
|
7
|
Nolan RP, Printz MA. Modeling the subcutaneous pharmacokinetics of antibodies co-administered with rHuPH20. Clin Transl Sci 2024; 17:e13788. [PMID: 38561908 PMCID: PMC10985223 DOI: 10.1111/cts.13788] [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: 02/05/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Predicting the subcutaneous (SC) pharmacokinetics (PK) of antibodies in humans is challenging, with clinical data currently being the only reliable data source for modeling SC absorption and bioavailability. Recombinant human hyaluronidase PH20 (rHuPH20) is an enzyme that facilitates SC delivery of high-dose, high-volume therapeutics. Numerous monoclonal antibodies have been co-administered SC with rHuPH20 in a clinical setting, establishing an extensive PK database. The goal of this work is to demonstrate how aggregated clinical data can be leveraged in a universal modeling framework for characterizing SC antibody PK, resulting in parameterization that can be used in predictive simulations of new antibodies. Data for 10 individual antibodies co-administered SC with rHuPH20 were obtained from publicly available sources. PK modeling of each antibody was conducted using the same model structure, but uniquely parameterized. The model structure consisted of a two-compartment model to capture linear kinetics, plus a target-binding mechanism to accommodate nonlinear kinetics driven by antibody-target complex formation and elimination. The clinical PK profiles for all antibodies were accurately described using the universal modeling framework. The SC PK parameters of absorption and bioavailability were consistent across the range of antibody and target properties evaluated. SC administration with rHuPH20 yielded a 30% increase in absorption rate on average and similar or better bioavailability. These parameter values can serve as initial conditions for model-based PK predictions for new antibodies co-administered SC with rHuPH20 to enable evaluation of optimal SC dose and schedule regimens prior to and during clinical development.
Collapse
|
8
|
Ait-Oudhia S, Wang YM, Dosne AG, Roy A, Jin JY, Shen J, Kagan L, Musuamba FT, Zhang L, Kijima S, Gastonguay MR, Ouellet D. Challenging the Norm: A Multidisciplinary Perspective on Intravenous to Subcutaneous Bridging Strategies for Biologics. Clin Pharmacol Ther 2024; 115:412-421. [PMID: 38069528 DOI: 10.1002/cpt.3133] [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: 09/22/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023]
Abstract
The transition from intravenous (i.v.) to subcutaneous (s.c.) administration of biologics is a critical strategy in drug development aimed at improving patient convenience, compliance, and therapeutic outcomes. Focusing on the increasing role of model-informed drug development (MIDD) in the acceleration of this transition, an in-depth overview of the essential clinical pharmacology, and regulatory considerations for successful i.v. to s.c. bridging for biologics after the i.v. formulation has been approved are presented. Considerations encompass multiple aspects beginning with adequate pharmacokinetic (PK) and pharmacodynamic (i.e., exposure-response) evaluations which play a vital role in establishing comparability between the i.v. and s.c. routes of administrations. Selected key recommendations and points to consider include: (i) PK characterization of the s.c. formulation, supported by the increasing preclinical understanding of the s.c. absorption, and robust PK study design and analyses in humans; (ii) a thorough characterization of the exposure-response profiles including important metrics of exposure for both efficacy and safety; (iii) comparability studies designed to meet regulatory considerations and support approval of the s.c. formulation, including noninferiority studies with PK and/or efficacy and safety as primary end points; and (iv) comprehensive safety package addressing assessments of immunogenicity and patients' safety profile with the new route of administration. Recommendations for successful bridging strategies are evolving and MIDD approaches have been used successfully to accelerate the transition to s.c. dosing, ultimately leading to improved patient experiences, adherence, and clinical outcomes.
Collapse
Affiliation(s)
| | - Yow-Ming Wang
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anne-Gaelle Dosne
- Janssen Research & Development, LLC, Beerse, Belgium
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Amit Roy
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jin Y Jin
- Genentech Inc., South San Francisco, California, USA
| | - Jun Shen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Leonid Kagan
- Department of Pharmaceutics and Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Flora T Musuamba
- Belgian Federal Agency for Medicines and Health Products, Brussels, Belgium
- NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Lucia Zhang
- Health Canada, Biologic and Radiopharmaceutical Drugs Directorate, Ottawa, Ontario, Canada
| | - Shinichi Kijima
- Pharmaceuticals and Medical Devices Agency (PMDA), Tokyo, Japan
| | | | - Daniele Ouellet
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| |
Collapse
|
9
|
Guo Y, Remaily BC, Thomas J, Kim K, Kulp SK, Mace TA, Ganesan LP, Owen DH, Coss CC, Phelps MA. Antibody Drug Clearance: An Underexplored Marker of Outcomes with Checkpoint Inhibitors. Clin Cancer Res 2024; 30:942-958. [PMID: 37921739 PMCID: PMC10922515 DOI: 10.1158/1078-0432.ccr-23-1683] [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: 06/13/2023] [Revised: 08/23/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Immune-checkpoint inhibitor (ICI) therapy has dramatically changed the clinical landscape for several cancers, and ICI use continues to expand across many cancer types. Low baseline clearance (CL) and/or a large reduction of CL during treatment correlates with better clinical response and longer survival. Similar phenomena have also been reported with other monoclonal antibodies (mAb) in cancer and other diseases, highlighting a characteristic of mAb clinical pharmacology that is potentially shared among various mAbs and diseases. Though tempting to attribute poor outcomes to low drug exposure and arguably low target engagement due to high CL, such speculation is not supported by the relatively flat exposure-response relationship of most ICIs, where a higher dose or exposure is not likely to provide additional benefit. Instead, an elevated and/or increasing CL could be a surrogate marker of the inherent resistant phenotype that cannot be reversed by maximizing drug exposure. The mechanisms connecting ICI clearance, therapeutic efficacy, and resistance are unclear and likely to be multifactorial. Therefore, to explore the potential of ICI CL as an early marker for efficacy, this review highlights the similarities and differences of CL characteristics and CL-response relationships for all FDA-approved ICIs, and we compare and contrast these to selected non-ICI mAbs. We also discuss underlying mechanisms that potentially link mAb CL with efficacy and highlight existing knowledge gaps and future directions where more clinical and preclinical investigations are warranted to clearly understand the value of baseline and/or time-varying CL in predicting response to ICI-based therapeutics.
Collapse
Affiliation(s)
- Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Bryan C. Remaily
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Justin Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Kyeongmin Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Samuel K. Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Latha P. Ganesan
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Dwight H. Owen
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Mitch A. Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| |
Collapse
|
10
|
Campbell E, Adamson H, Luxton T, Tiede C, Wälti C, Tomlinson DC, Jeuken LJC. Therapeutic drug monitoring of immunotherapies with novel Affimer-NanoBiT sensor construct. SENSORS & DIAGNOSTICS 2024; 3:104-111. [PMID: 38249540 PMCID: PMC10795742 DOI: 10.1039/d3sd00126a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
Concentration-therapeutic efficacy relationships have been observed for several therapeutic monoclonal antibodies (TmAb), where low circulating levels can result in ineffective treatment and high concentrations can cause adverse reactions. Rapid therapeutic drug monitoring (TDM) of TmAb drugs would provide the opportunity to adjust an individual patient's dosing regimen to improve treatment results. However, TDM for immunotherapies is currently limited to centralised testing methods with long sample-collection to result timeframes. Here, we show four point-of-care (PoC) TmAb biosensors by combining anti-idiotypic Affimer proteins and NanoBiT split luciferase technology at a molecular level to provide a platform for rapid quantification (<10 minutes) for four clinically relevant TmAb (rituximab, adalimumab, ipilimumab and trastuzumab). The rituximab sensor performed best with 4 pM limit of detection (LoD) and a quantifiable range between 8 pM-2 nM with neglectable matrix effects in serum up to 1%. After dilution of serum samples, the resulting quantifiable range for all four sensors falls within the clinically relevant range and compares favourably with the sensitivity and/or time-to-result of current ELISA standards. Further development of these sensors into a PoC test may improve treatment outcome and quality of life for patients receiving immunotherapy.
Collapse
Affiliation(s)
- Emma Campbell
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
| | - Hope Adamson
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
| | - Timothy Luxton
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
| | - Christian Tiede
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
- School of Molecular and Cellular Biology, University of Leeds Leeds LS2 9JT UK
| | - Christoph Wälti
- School of Electronic and Electrical Engineering, University of Leeds LS2 9JT UK
| | - Darren C Tomlinson
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
- School of Molecular and Cellular Biology, University of Leeds Leeds LS2 9JT UK
| | - Lars J C Jeuken
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
- Leiden Institute of Chemistry, Leiden University PO Box 9502 2300 RA Leiden The Netherlands
| |
Collapse
|
11
|
Reddy N, Reddy P, Ranpura A, Maharaj N, Arora R, Mamillapalli G, Adhav AS, Diwan AK, Manikhas A, Krasnozhon D. Efficacy, Safety, Pharmacokinetics, and Immunogenicity of DRL-Trastuzumab Versus Herceptin in Human Epidermal Growth Factor Receptor 2-Positive Metastatic Breast Cancer: A Randomized Controlled Trial. JCO Glob Oncol 2024; 10:e2200328. [PMID: 38237093 PMCID: PMC10805439 DOI: 10.1200/go.22.00328] [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: 09/30/2022] [Revised: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 01/23/2024] Open
Abstract
PURPOSE Dr Reddy's Laboratories Trastuzumab (DRL_TZ) is a biosimilar to Herceptin under development. The present study was conducted to evaluate efficacy, safety, pharmacokinetics (PKs), and immunogenicity of DRL_TZ in comparison with the reference medicinal product (RMP) along with concomitant weekly paclitaxel in patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer (MBC). METHODS This was a randomized, double-blind study in female patients with HER2-positive MBC, randomly assigned in a 1:1 ratio to receive either DRL_TZ or the RMP, that is, an innovator product sourced from the European region, along with additional chemotherapy, as first-line treatment for up to 24 weeks. The primary end point was the best overall response rate (ORR) as per RECIST 1.1 criteria. Progression-free survival rate at 6 months (PFS6), safety, immunogenicity, and PK parameters were assessed as secondary end points. RESULTS A total of 164 patients were randomly assigned to receive either DRL_TZ or the RMP. Best ORR in the per-protocol population was comparable, 91.9% (93.3% CI, 83.2 to 96.3) versus 82.1% (93.3% CI, 72.0 to 89.1) in DRL_TZ and RMP arms, respectively; the difference between the arms was 9.8% with a 93.3% CI of -1.3 to 20.8. The PFS6 rate, safety, PK profile, and antidrug antibody incidence were comparable. An additional 44 patients were recruited in the postrandomization phase, in an open-label manner, and started on DRL_TZ to generate more data on efficacy, safety, and immunogenicity. The additional data with DRL_TZ, when pooled, were similar to the RMP data. CONCLUSION DRL_TZ was found to have similar efficacy and comparable safety, PK, and immunogenicity profiles as the RMP.
Collapse
Affiliation(s)
| | | | | | | | - Rajendersingh Arora
- Sujan Surgical Cancer Hospital and Amravati Cancer Foundation, Amravati, India
| | | | | | - Ashok Kumar Diwan
- Government Medical College and Hospital, Medical College Square, Nagpur, India
| | - Alexey Manikhas
- St Petersburg State Budget Healthcare Institution “City Clinical Oncology Center”, St Petersburg, Russia
| | - Dmitriy Krasnozhon
- State Budget Healthcare Institution “Leningrad Regional Oncology Center”, St Petersburg, Russia
| |
Collapse
|
12
|
Bittner B, Schmidt J. Advancing Subcutaneous Dosing Regimens for Biotherapeutics: Clinical Strategies for Expedited Market Access. BioDrugs 2024; 38:23-46. [PMID: 37831325 PMCID: PMC10789662 DOI: 10.1007/s40259-023-00626-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2023] [Indexed: 10/14/2023]
Abstract
In recent years, subcutaneous administration of biotherapeutics has made significant progress. The self-administration market for rheumatoid arthritis has witnessed the introduction of additional follow-on biologics, while the first subcutaneous dosing options for monoclonal antibodies have become available for multiple sclerosis. Oncology has also seen advancements with the authorization of high-volume subcutaneous formulations, facilitated by the development of high-concentration formulations and innovative delivery systems. Regulatory and Health Technology Assessment bodies increasingly consider preference data in filing dossiers, particularly in evaluating novel drug delivery methods. The adoption of a pharmacokinetic-based clinical bridging approach has become standard for transitioning from intravenous to subcutaneous administration. Non-inferiority studies with pharmacokinetics as the only primary endpoint have started deviating from traditional randomization schemes, favoring the subcutaneous route and comparing with historical intravenous data. While nonclinical and computational models made progress in predicting safety and immunogenicity for subcutaneously dosed antibodies, clinical trial evidence remains essential due to inter-individual variations and the impact of formulation parameters on anti-drug antibody formation. Ongoing technological advancements and the expanding knowledge base on pharmacokinetic-pharmacodynamic correlation across specialty areas are expected to further accelerate clinical development of subcutaneous biologics.
Collapse
Affiliation(s)
- Beate Bittner
- Global Product Strategy, Product Optimization, F. Hoffmann-La Roche, Grenzacher Strasse 124, 4070, Basel, Switzerland.
| | - Johannes Schmidt
- Global Product Strategy, Product Optimization, F. Hoffmann-La Roche, Grenzacher Strasse 124, 4070, Basel, Switzerland
| |
Collapse
|
13
|
Campbell E, Adamson H, Kohl D, Tiede C, Wälti C, Tomlinson DC, Jeuken LJC. Enzyme - Switch sensors for therapeutic drug monitoring of immunotherapies. Biosens Bioelectron 2023; 237:115488. [PMID: 37419072 PMCID: PMC10427837 DOI: 10.1016/j.bios.2023.115488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/19/2023] [Accepted: 06/16/2023] [Indexed: 07/09/2023]
Abstract
Therapeutic monoclonal antibodies (TmAb) have emerged as effective treatments for a number of cancers and autoimmune diseases. However, large interpatient disparities in the pharmacokinetics of TmAb treatment requires close therapeutic drug monitoring (TDM) to optimise dosage for individual patients. Here we demonstrate an approach for achieving rapid, sensitive quantification of two monoclonal antibody therapies using a previously described enzyme switch sensor platform. The enzyme switch sensor consists of a β-lactamase - β-lactamase inhibitor protein (BLA-BLIP) complex with two anti-idiotype binding proteins (Affimer proteins) as recognition elements. The BLA-BLIP sensor was engineered to detect two TmAbs (trastuzumab and ipilimumab) by developing constructs incorporating novel synthetic binding reagents to each of these mAbs. Trastuzumab and ipilimumab were successfully monitored with sub nM sensitivity in up to 1% serum, thus covering the relevant therapeutic range. Despite the modular design, the BLA-BLIP sensor was unsuccessful in detecting two further TmAbs (rituximab and adalimumab), an explanation for which was explored. In conclusion, the BLA-BLIP sensors provide a rapid biosensor for TDM of trastuzumab and ipilimumab with the potential to improve therapy. The sensitivity of this platform alongside its rapid action would be suitable for bedside monitoring in a point-of-care (PoC) setting.
Collapse
Affiliation(s)
- Emma Campbell
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom
| | - Hope Adamson
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom
| | - Declan Kohl
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom
| | - Christian Tiede
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Christoph Wälti
- School of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Darren C Tomlinson
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Lars J C Jeuken
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom; Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA, Leiden, the Netherlands.
| |
Collapse
|
14
|
Homšek A, Spasić J, Nikolić N, Stanojković T, Jovanović M, Miljković B, Vučićević KM. Pharmacokinetic characterization, benefits and barriers of subcutaneous administration of monoclonal antibodies in oncology. J Oncol Pharm Pract 2023; 29:431-440. [PMID: 36349366 DOI: 10.1177/10781552221137702] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Therapeutic monoclonal antibodies in oncology are slowly becoming the dominant treatment option for many different cancer types. The main route of administration, infusion, requires extensive product preparations, patient hospitalization and close monitoring. Patient comfort improvement, staff workload reduction and cost savings dictated the development of subcutaneous formulations. The aim of this review is to present pharmacokinetic characteristics of subcutaneous products, discuss the differences between intravenous and subcutaneous routes and to point out the advantages as well as challenges of administration route shift from the formulation development and pharmacometric angle. DATA SOURCES Food and Drug administration's Purple book database and electronic medicines compendium were used to identify monoclonal antibodies in oncology approved as subcutaneous forms. Using keywords subcutaneous, monoclonal antibodies, pharmacokinetics, model, as well as specific drugs previously identified, both PubMed and ScienceDirect databases were researched. DATA SUMMARY There are currently six approved subcutaneous onco-monoclonal antibodies on the market. For each of them, exposure to the drug was similar in relation to infusion, treatment effectiveness was the same, administration was well tolerated by the patients and costs of the medical service were reduced. CONCLUSION Development of subcutaneous forms for existing and emerging new monoclonal antibodies for cancer treatment as well as shifting from administration via infusion should be encouraged due to patient preference, lower costs and overall lack of substantial differences in efficacy and safety between the two routes.
Collapse
Affiliation(s)
- Ana Homšek
- Department of Pharmacokinetics and Clinical Pharmacy, 186111University of Belgrade - Faculty of Pharmacy, Belgrade, Republic of Serbia
| | - Jelena Spasić
- Clinic for Medical Oncology, 119083Institute for Oncology and Radiology of Serbia, Belgrade, Republic of Serbia
| | - Neda Nikolić
- Clinic for Medical Oncology, 119083Institute for Oncology and Radiology of Serbia, Belgrade, Republic of Serbia
| | - Tatjana Stanojković
- Department of Experimental Oncology, 119083Institute for Oncology and Radiology of Serbia, Belgrade, Republic of Serbia
| | - Marija Jovanović
- Department of Pharmacokinetics and Clinical Pharmacy, 186111University of Belgrade - Faculty of Pharmacy, Belgrade, Republic of Serbia
| | - Branislava Miljković
- Department of Pharmacokinetics and Clinical Pharmacy, 186111University of Belgrade - Faculty of Pharmacy, Belgrade, Republic of Serbia
| | - Katarina M Vučićević
- Department of Pharmacokinetics and Clinical Pharmacy, 186111University of Belgrade - Faculty of Pharmacy, Belgrade, Republic of Serbia
| |
Collapse
|
15
|
Soltantabar P, Lon HK, Parivar K, Wang DD, Elmeliegy M. Optimizing benefit/risk in oncology: Review of post-marketing dose optimization and reflections on the road ahead. Crit Rev Oncol Hematol 2023; 182:103913. [PMID: 36681205 DOI: 10.1016/j.critrevonc.2023.103913] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Oncology therapies shifted from chemotherapy to molecularly targeted agents and finally to the era of immune-oncology agents. In contrast to cytotoxic agents, molecularly targeted agents are more selective, exhibit a wider therapeutic window, and may maximally modulate tumor growth at doses lower than the maximum tolerated dose (MTD). However, first-in-patient oncology studies for molecularly targeted agents continued to evaluate escalating doses using limited number of patients per dose cohort assessing dose-limiting toxicities to identify the MTD which is commonly selected for further development adopting a 'more is better' approach that led to several post-marketing requirement (PMR) studies to evaluate alternative, typically lower, doses or dosing frequencies to optimize the benefit-risk profile. In this review, post-marketing dose optimization efforts were reviewed including those required by a regulatory pathway or voluntarily conducted by the sponsor to improve efficacy, safety, or method of administration. Lessons learned and future implications from this deep dive review are discussed considering the evolving regulatory landscape on dose optimization for oncology compounds.
Collapse
Affiliation(s)
| | - Hoi-Kei Lon
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | | - Diane D Wang
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | |
Collapse
|
16
|
Desai M, Kundu A, Hageman M, Lou H, Boisvert D. Monoclonal antibody and protein therapeutic formulations for subcutaneous delivery: high-concentration, low-volume vs. low-concentration, high-volume. MAbs 2023; 15:2285277. [PMID: 38013454 DOI: 10.1080/19420862.2023.2285277] [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: 10/05/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
Biologic drugs are used to treat a variety of cancers and chronic diseases. While most of these treatments are administered intravenously by trained healthcare professionals, a noticeable trend has emerged favoring subcutaneous (SC) administration. SC administration of biologics poses several challenges. Biologic drugs often require higher doses for optimal efficacy, surpassing the low volume capacity of traditional SC delivery methods like autoinjectors. Consequently, high concentrations of active ingredients are needed, creating time-consuming formulation obstacles. Alternatives to traditional SC delivery systems are therefore needed to support higher-volume biologic formulations and to reduce development time and other risks associated with high-concentration biologic formulations. Here, we outline key considerations for SC biologic drug formulations and delivery and explore a paradigm shift: the flexibility afforded by low-to-moderate-concentration drugs in high-volume formulations as an alternative to the traditionally difficult approach of high-concentration, low-volume SC formulation delivery.
Collapse
Affiliation(s)
- M Desai
- Medical Affairs, Enable Injections, Inc, Cincinnati, OH, USA
| | - A Kundu
- Manufacturing Sciences, Takeda Pharmaceuticals, Brooklyn Park, MN, USA
| | - M Hageman
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| | - H Lou
- Biopharmaceutical Innovation & Optimization Center, The University of Kansas, Lawrence, KS, USA
| | - D Boisvert
- Independent Chemistry Manufacturing & Controls (CMC) Consultant, El Cerrito, CA, USA
| |
Collapse
|
17
|
Alternative trastuzumab dosing strategies in HER2-positive early breast cancer are associated with patient out-of-pocket savings. NPJ Breast Cancer 2022; 8:32. [PMID: 35288585 PMCID: PMC8921207 DOI: 10.1038/s41523-022-00393-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 02/02/2022] [Indexed: 11/08/2022] Open
Abstract
AbstractPatients with breast cancer frequently experience financial hardship, often due to the high costs of anti-cancer drugs. We sought to develop alternative trastuzumab dosing strategies, compare their pharmacokinetic effectiveness to standard dosing, and assess the expected financial implications of transitioning to them. We extracted clinical data from the records of 135 retrospectively identified patients with HER2-positive early breast cancer at a single, urban comprehensive cancer center who were treated with trastuzumab between 2017 and 2019. We performed pharmacokinetic simulations on a range of trastuzumab dose levels and frequencies, assessing efficacy by trough trastuzumab concentration (Ctrough) and population and individual likelihoods of Ctrough exceeding trastuzumab minimum effective concentration (MEC). We performed deterministic financial modeling to estimate the treatment-associated financial savings from alternative dosing strategies. Trastuzumab maintenance doses of 4 mg/kg every 3 weeks (Q3W) and 6 mg/kg every 4 weeks (Q4W) had nearly identical probabilities of Ctrough being above MEC as standard of care 6 mg/kg every 3 weeks. In the primary financial analysis, both trastuzumab 4 mg/kg Q3W and 6 mg/kg Q4W were associated with significant drug- and administration-related out-of-pocket cost savings over the duration of therapy, ranging from $765 (neoadjuvant, Q4W) to $2791 (adjuvant, Q4W). In particular, Q4W trastuzumab increased savings related to lost wages and travel cost avoidance. Low-dose and reduced frequency trastuzumab in appropriately selected patients may significantly reduce total drug utilization and meaningfully reduce patient financial toxicity. Prospective clinical trials evaluating low-dose or reduced-frequency administration of therapeutic monoclonal antibodies are warranted and needed.
Collapse
|
18
|
Al Ojaimi Y, Blin T, Lamamy J, Gracia M, Pitiot A, Denevault-Sabourin C, Joubert N, Pouget JP, Gouilleux-Gruart V, Heuzé-Vourc'h N, Lanznaster D, Poty S, Sécher T. Therapeutic antibodies - natural and pathological barriers and strategies to overcome them. Pharmacol Ther 2021; 233:108022. [PMID: 34687769 PMCID: PMC8527648 DOI: 10.1016/j.pharmthera.2021.108022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023]
Abstract
Antibody-based therapeutics have become a major class of therapeutics with over 120 recombinant antibodies approved or under review in the EU or US. This therapeutic class has experienced a remarkable expansion with an expected acceleration in 2021-2022 due to the extraordinary global response to SARS-CoV2 pandemic and the public disclosure of over a hundred anti-SARS-CoV2 antibodies. Mainly delivered intravenously, alternative delivery routes have emerged to improve antibody therapeutic index and patient comfort. A major hurdle for antibody delivery and efficacy as well as the development of alternative administration routes, is to understand the different natural and pathological barriers that antibodies face as soon as they enter the body up to the moment they bind to their target antigen. In this review, we discuss the well-known and more under-investigated extracellular and cellular barriers faced by antibodies. We also discuss some of the strategies developed in the recent years to overcome these barriers and increase antibody delivery to its site of action. A better understanding of the biological barriers that antibodies have to face will allow the optimization of antibody delivery near its target. This opens the way to the development of improved therapy with less systemic side effects and increased patients' adherence to the treatment.
Collapse
Affiliation(s)
- Yara Al Ojaimi
- UMR 1253, iBrain, Inserm, 37000 Tours, France; University of Tours, 37000 Tours, France
| | - Timothée Blin
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| | - Juliette Lamamy
- University of Tours, 37000 Tours, France; GICC, EA7501, 37000 Tours, France
| | - Matthieu Gracia
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | - Aubin Pitiot
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| | | | - Nicolas Joubert
- University of Tours, 37000 Tours, France; GICC, EA7501, 37000 Tours, France
| | - Jean-Pierre Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | | | | | - Débora Lanznaster
- UMR 1253, iBrain, Inserm, 37000 Tours, France; University of Tours, 37000 Tours, France
| | - Sophie Poty
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier F-34298, France
| | - Thomas Sécher
- University of Tours, 37000 Tours, France; UMR 1100, CEPR, Inserm, 37000 Tours, France
| |
Collapse
|
19
|
Wang B, Deng R, Hennig S, Badovinac Crnjevic T, Kaewphluk M, Kågedal M, Quartino AL, Girish S, Li C, Kirschbrown WP. Population pharmacokinetic and exploratory exposure-response analysis of the fixed-dose combination of pertuzumab and trastuzumab for subcutaneous injection in patients with HER2-positive early breast cancer in the FeDeriCa study. Cancer Chemother Pharmacol 2021; 88:499-512. [PMID: 34106303 PMCID: PMC8187458 DOI: 10.1007/s00280-021-04296-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/15/2021] [Indexed: 11/30/2022]
Abstract
Purpose To characterize pertuzumab pharmacokinetics (PK) in FeDeriCa (NCT03493854: fixed-dose combination of pertuzumab and trastuzumab for subcutaneous injection [PH FDC SC] versus intravenous pertuzumab plus trastuzumab); derive individual pertuzumab exposures in the PH FDC SC arm for subsequent pertuzumab exposure–response (ER) analyses; compare observed trastuzumab PK with predicted exposures from a previous SC trastuzumab model; assess whether pertuzumab affects trastuzumab PK; evaluate pertuzumab exposure–efficacy and –safety relationships and support the approved SC dosing regimen. Methods Population pharmacokinetic modeling and simulations were used to describe the data. Standard goodness-of-fit diagnostics and prediction-corrected visual predictive checks were used for model performance assessment. Covariates were included from previously reported models. ER analysis was conducted using logistic regression. Results SC pertuzumab PK was described adequately by a two-compartment model with first-order absorption; significant covariates included in the final model were albumin, lean body weight, and Asian region; however, these appeared not to be clinically relevant. Trastuzumab concentrations were described adequately by the previous model; there was no evidence of a pertuzumab effect on trastuzumab PK as part of PH FDC SC and higher model-predicted pertuzumab exposure was not associated with differences in pathologic complete response rate or an increased probability of selected grade ≥ 3 adverse events of interest. Conclusion The approved PH FDC SC dose [loading: 1200/600 mg pertuzumab/trastuzumab (15 mL); maintenance: 600 mg pertuzumab/trastuzumab (10 mL) and 2000 U/mL recombinant human hyaluronidase every 3 weeks] provides a positive benefit–risk profile with comparable efficacy and safety to intravenous pertuzumab plus trastuzumab. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-021-04296-0.
Collapse
Affiliation(s)
- Bei Wang
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Rong Deng
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | | | | | | | - Matts Kågedal
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Angelica L Quartino
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
- Clinical Pharmacology and Quantitative Pharmacology, AstraZeneca, Gothenburg, Sweden
| | - Sandhya Girish
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
- Gilead Sciences, Inc., Foster City, CA, USA
| | - Chunze Li
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | | |
Collapse
|
20
|
Kolberg HC, Jackisch C, Hurvitz SA, Winstone J, Barham H, Hanes V, Courmier D. Is weight-based IV dosing of trastuzumab preferable to SC fixed-dose in some patients? A systematic scoping review. Breast 2021; 57:95-103. [PMID: 33799233 PMCID: PMC8044716 DOI: 10.1016/j.breast.2021.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/11/2021] [Accepted: 03/14/2021] [Indexed: 12/16/2022] Open
Abstract
Trastuzumab, a key treatment for HER2-positive breast cancer, is available in weight-based IV and fixed-dose (600 mg) SC formulations. While the Phase 3 HannaH trial indicated non-inferiority of the SC formulation, there is some concern that the target plasma concentration may not be reached in overweight/obese patients whereas low-body-weight patients may be at risk of toxicity. This scoping review evaluated whether overweight/obese patients are at risk of below-target exposure with fixed-dose SC trastuzumab, whether low-body-weight patients are at risk of increased toxicity, especially cardiotoxicity, and whether IV and SC trastuzumab are equivalent in terms of treatment-emergent adverse events (TEAEs) (e.g. infections). Thirty-seven publications that met the eligibility criteria were included. Body weight is not an important determinant of exposure to trastuzumab at steady state (i.e. pre-dose cycle 8); however, real-world evidence suggests that the target concentration (20 μg/mL) may not be reached with the first SC dose in overweight/obese patients. There is no evidence that low-body-weight patients are at increased risk of cardiotoxicity with SC trastuzumab, although this may be confounded by the higher rate of cardiovascular comorbidities in overweight patients. In Phase 3 trials, SC trastuzumab was associated with higher rates of ISRs, ADAs and SAEs, the latter often requiring hospitalization and occurring during adjuvant treatment when patients are not burdened by chemotherapy. The route of administration of trastuzumab (IV vs SC) in different treatment settings should be discussed with the patient, taking into account the risks and benefits associated with each route.
Collapse
Affiliation(s)
- Hans-Christian Kolberg
- Department of Gynecology and Obstetrics, Breast and Gynecologic Cancer Center, Marienhospital, Josef-Albers-Str. 70, 46236, Bottrop, Germany.
| | - Christian Jackisch
- Department of Obstetrics and Gynecology, Breast and Gynecologic Cancer Center, Sana Klinikum Offenbach, Starkenburgring 66, D-63069, Offenbach, Germany.
| | - Sara A Hurvitz
- Department of Medicine, Division of Hematology/Oncology, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Julie Winstone
- Perscribo Medical Communications Ltd, 7 York Close, Kempshott Rise, Basingstoke, RG22 4PU, UK.
| | - Helen Barham
- Perscribo Medical Communications Ltd, 7 York Close, Kempshott Rise, Basingstoke, RG22 4PU, UK
| | - Vladimir Hanes
- Amgen Biosimilar Business Unit, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320USA, USA.
| | - Delphine Courmier
- Amgen Global Health Economics, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320USA, USA.
| |
Collapse
|
21
|
Solans BP, Garrido MJ, Trocóniz IF. Drug Exposure to Establish Pharmacokinetic-Response Relationships in Oncology. Clin Pharmacokinet 2021; 59:123-135. [PMID: 31654368 DOI: 10.1007/s40262-019-00828-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In the oncology field, understanding the relationship between the dose administered and the exerted effect is particularly important because of the narrow therapeutic index associated with anti-cancer drugs and the high interpatient variability. Therefore, in this review, we provide a critical perspective of the different methods of characterising treatment exposure in the oncology setting. The increasing number of modelling applications in oncology reflects the applicability and the impact of pharmacometrics on all phases of the drug development process and patient management as well. Pharmacometric modelling is a worthy component within the current paradigm of model-based drug development, but pharmacometric modelling techniques are also accessible for the clinician in the optimisation of current oncology therapies. Consequently, the application of population models in a hospital setting by generating close collaborations between physicians and pharmacometricians is highly recommended, providing a systematic means of developing and assessing model-based metrics as 'drivers' for various responses to treatments, which can then be evaluated as predictors for treatment success. Characterising the key determinants of variability in exposure is of particular importance for anticancer agents, as efficacy and toxicity are associated with exposure. We present the different strategies to describe and predict drug exposure that can be applied depending on the data available, with the objective of obtaining the most useful information in the patients' favour throughout the full drug cycle. Therefore, the objective of the present article is to review the different approaches used to characterise a patient's exposure to oncology drugs, which will result in a better understanding of the time course of the response and the magnitude of interpatient variability.
Collapse
Affiliation(s)
- Belén P Solans
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, C/Irunlarrea s/n, 31008, Pamplona, Navarra, Spain. .,Navarra Institute for Health Research (IdisNA), University of Navarra, Pamplona, Spain.
| | - María Jesús Garrido
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, C/Irunlarrea s/n, 31008, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IdisNA), University of Navarra, Pamplona, Spain
| | - Iñaki F Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, C/Irunlarrea s/n, 31008, Pamplona, Navarra, Spain. .,Navarra Institute for Health Research (IdisNA), University of Navarra, Pamplona, Spain.
| |
Collapse
|
22
|
Simulating Costs of Intravenous Biosimilar Trastuzumab vs. Subcutaneous Reference Trastuzumab in Adjuvant HER2-Positive Breast Cancer: A Belgian Case Study. Pharmaceuticals (Basel) 2021; 14:ph14050450. [PMID: 34064559 PMCID: PMC8151172 DOI: 10.3390/ph14050450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 11/17/2022] Open
Abstract
This study aimed to compare drug costs and healthcare costs of a 1 year adjuvant course with intravenous biosimilar trastuzumab vs. subcutaneous reference trastuzumab in HER2-positive breast cancer from the Belgian hospital perspective. Our simulation is based on the methodology used by Tjalma and colleagues, and considered costs of drugs, healthcare professional time and consumables. We calculated intravenous drug costs for different body weights, and computed drug costs and healthcare costs to treat 100 patients with either trastuzumab formulation, assuming a binomial body weight distribution in this sample. Scenarios were run to account for drug discounts and intravenous vial sharing. Drug costs amounted to €1,431,282 with intravenous biosimilar trastuzumab and €1,522,809 with subcutaneous reference trastuzumab for a sample of 100 patients in the base case analysis. When healthcare professional time and consumables were also considered, healthcare costs with intravenous biosimilar trastuzumab were similar to those with subcutaneous reference trastuzumab. Differences in healthcare costs between intravenous biosimilar trastuzumab and subcutaneous reference trastuzumab depended on the level of discounts on these formulations and on intravenous vial sharing. Our case study demonstrates that comparing costs of intravenous vs. subcutaneous formulations is complex and multifactorial, and entails more than a simple cost comparison of products.
Collapse
|
23
|
Siri-Angkul N, Chattipakorn SC, Chattipakorn N. The mechanistic insights of the arrhythmogenic effect of trastuzumab. Biomed Pharmacother 2021; 139:111620. [PMID: 33901874 DOI: 10.1016/j.biopha.2021.111620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases and cancers are the leading causes of deaths globally, and an increasing proportion of cancer patients is suffering from cardiac adverse effects of chemotherapeutic drugs. Trastuzumab, a monoclonal antibody that inhibits the activity of the human epidermal growth factor receptor 2 (HER2), is a potent targeted therapy for HER2-positive malignancies. Despite the impressive antineoplastic efficacy, the cardiotoxicity of trastuzumab frequently limits its use. Trastuzumab-induced cardiac contractile dysfunction has been extensively studied, yet the electrophysiological side effect of trastuzumab remains poorly characterized. Growing evidence from basic and clinical studies supports the link between trastuzumab treatment and arrhythmias. This review comprehensively summarizes relevant information from those reports, discusses their limitations, and suggests future research directions. We aim to encourage further investigations that will provide valuable insights to devise cardioprotective strategies against trastuzumab-induced cardiotoxicity.
Collapse
Affiliation(s)
- Natthaphat Siri-Angkul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
24
|
Wiedermann U, Garner-Spitzer E, Chao Y, Maglakelidze M, Bulat I, Dechaphunkul A, Arpornwirat W, Charoentum C, Yen CJ, Yau TC, Tanasanvimon S, Maneechavakajorn J, Sookprasert A, Bai LY, Chou WC, Ungtrakul T, Drinic M, Tobias J, Zielinski CC, Chong L, Ede NJ, Marino MT, Good AJ. Clinical and Immunologic Responses to a B-Cell Epitope Vaccine in Patients with HER2/neu-Overexpressing Advanced Gastric Cancer-Results from Phase Ib Trial IMU.ACS.001. Clin Cancer Res 2021; 27:3649-3660. [PMID: 33879458 DOI: 10.1158/1078-0432.ccr-20-3742] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/30/2020] [Accepted: 04/16/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE HER2/neu is overexpressed in up to 30% of gastroesophageal adenocarcinomas (GEA) and linked to poor prognosis. Recombinant mAbs to treat HER2/neu-overexpressing cancers are effective with limitations, including resistance and toxicity. Therefore, we developed a therapeutic B-cell epitope vaccine (IMU-131/HER-Vaxx) consisting of three fused B-cell epitopes from the HER2/neu extracellular domain coupled to CRM197 and adjuvanted with Montanide. This phase Ib study aimed to evaluate the optimal/safe dose leading to immunogenicity and clinical responses (https//clinicaltrials.gov/ct2/show/NCT02795988). PATIENTS AND METHODS A total of 14 patients with HER2/neu-overexpressing GEA were enrolled, and dose escalation (10, 30, 50 μg) was performed in three cohorts (C). Immunogenicity was evaluated by HER2-specific Abs and cellular responses, clinical responses by CT scans according to RECIST version 1.1. RESULTS IMU-131 was safe without vaccine-related significant local/systemic reactions or serious adverse events. A total of 11 of 14 patients were evaluable for changes in tumor size and vaccine-specific immune responses. One patient showed complete, 5 partial responses, and 4 stable diseases as their best response. HER2-specific IgG levels were dose dependent. In contrast to patients in C1 and C2, all patients in C3 mounted substantial HER2-specific Ab levels. In addition, cellular vaccine responses, such as Th1-biased cytokine ratios and reduced regulatory T cell numbers, were generated. Progression-free survival was prolonged in C3, correlating with the vaccine-specific humoral and cellular responses. CONCLUSIONS IMU-131 was well tolerated and safe. The induced HER2-specific Abs and cellular responses were dose dependent and correlated with clinical responses. The highest dose (50 μg) was recommended for further evaluation in a phase II trial, with chemotherapy + IMU-131 or chemotherapy alone, which is currently ongoing.
Collapse
Affiliation(s)
- Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - Erika Garner-Spitzer
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Yee Chao
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Iurie Bulat
- ARENSIA Exploratory Medicine Research Unit, Institute of Oncology, Chisinau, Republic of Moldova
| | - Arunee Dechaphunkul
- Department of Medicine, Songklanagarind Hospital, Prince of Songkla University, Hat Yai, Thailand
| | | | - Chaiyut Charoentum
- Maharaj Nakorn Chiang Mai Hospital, Mueang Chiang Mai District, Thailand
| | | | - Thomas Cheung Yau
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong
| | | | | | | | - Li-Yuan Bai
- China Medical University Hospital, Taichung City, Taiwan
| | - Wen-Chi Chou
- Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Teerapat Ungtrakul
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Bangkok, Thailand
| | - Mirjana Drinic
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Joshua Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
25
|
Wardley A, Canon JL, Elsten L, Peña Murillo C, Badovinac Crnjevic T, Fredriksson J, Piccart M. Flexible care in breast cancer. ESMO Open 2021; 6:100007. [PMID: 33450658 PMCID: PMC7811121 DOI: 10.1016/j.esmoop.2020.100007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/03/2020] [Indexed: 12/30/2022] Open
Abstract
Treatment of patients with cancer in hospitals or clinics is resource-intensive and imposes a burden on patients. 'Flexible care' is a term that can be used to describe treatment administered outside the oncology ward, oncological outpatient clinic or office-based oncologist setting. Programmes that reduce travel burden by bringing cancer treatment to the patient's home, workplace or closer to the patient's home, in the form of satellite clinics or mobile cancer units, expand treatment capacity and are well received. Clinical trial data show that, compared with intravenous administration, subcutaneous (s.c.) administration of trastuzumab is preferred by patients with breast cancer (BC), saves healthcare professionals' (HCPs) time, reduces drug preparation and administration time and reduces direct and indirect costs. As such, s.c. trastuzumab is well suited to flexible care. The results of a Belgian study (BELIS) show that home administration of s.c. trastuzumab is feasible and preferred by patients with BC. Numerous programmes and pilot studies in Europe show that s.c. trastuzumab can be administered effectively in the patient's home, in primary care settings or local hospitals. Such programmes require planning, training, careful patient selection and technology to link patients, caregivers and specialists in oncology clinics. Once these elements are in place, flexible care offers patients with BC a choice of how treatment may be delivered and lead to improved quality of life, while reducing pressure on HCPs and hospitals. The concept of flexible care is particularly relevant amid the COVID-19 pandemic where guidelines have been developed encouraging remote care.
Collapse
Affiliation(s)
- A Wardley
- NIHR Manchester Clinical Research Facility at The Christie and Division of Cancer Sciences and University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - J-L Canon
- Service d'Oncologie-Hématologie, Site Notre-Dame, Grand Hôpital de Charleroi (GHdC), Charleroi, Belgium
| | - L Elsten
- Department of Medical Oncology, Amphia Hospital, Breda, The Netherlands
| | - C Peña Murillo
- Global Product Development, Medical Affairs, Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - J Fredriksson
- Global Product Development, Medical Affairs, Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - M Piccart
- Institut Jules Bordet, Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
26
|
Kokkali S, Ternant D, Kemmel V, Levêque D, Wendling F, Barthelemy P, Kurtz JE. Intravenous and subcutaneous administration of trastuzumab in a patient on peritoneal dialysis. Br J Clin Pharmacol 2021; 87:3372-3374. [PMID: 33464605 DOI: 10.1111/bcp.14731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- Stefania Kokkali
- Department of Medical Oncology, Strasbourg University Hospital, Strasbourg, France.,First Medical Oncology Department, Saint-Savvas Anticancer Hospital, Athens, Greece
| | - David Ternant
- Department of Medical Pharmacology, CHRU de Tours, Tours University Hospital, Tours, France
| | - Véronique Kemmel
- Biochemistry and Molecular Biology Department, Strasbourg University Hospital, Strasbourg, France
| | - Dominique Levêque
- Pharmacy Department, Strasbourg University Hospital, Strasbourg, France
| | - Fréderique Wendling
- Department of Medical Oncology, Strasbourg University Hospital, Strasbourg, France
| | - Philippe Barthelemy
- Department of Medical Oncology, Strasbourg University Hospital, Strasbourg, France
| | - Jean-Emmanuel Kurtz
- Department of Medical Oncology, Strasbourg University Hospital, Strasbourg, France
| |
Collapse
|
27
|
Dong W, Chen M, Wang J, Xia L, Wang Q, Nie X, Feng Y, Fang Y. rHuPH20-facilitated subcutaneous administration of monoclonal antibodies in cancer therapy. Immunotherapy 2020; 13:79-88. [PMID: 33198539 DOI: 10.2217/imt-2020-0204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: This meta-analysis aimed to evaluate the pharmacokinetics, efficacy, safety and immunogenicity of rHuPH20-facilitated subcutaneous (SC) administration of monoclonal antibody compared with intravenous (IV) administration for patients with cancer. Materials & methods: Outcomes included trough concentrations (Ctrough), overall response rate, adverse events, serious adverse events and antidrug antibody positivity rate. Subgroup analysis was also performed. Results: Five studies involving 1575 participants (788/787) were included. All studies met the non-inferiority criterion in Ctrough. No significant differences were observed in overall response rate (p = 0.12), adverse events (p = 0.05), and severe adverse events (p = 0.73) between SC and IV groups. The SC group also had lower immunogenicity than the IV group. Conclusion: rHuPH20-facilitated subcutaneous administration of monoclonal antibody is highly similar to IV administration in terms of pharmacokinetics, efficacy, and safety, but with lower immunogenicity.
Collapse
Affiliation(s)
- Wenliang Dong
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Min Chen
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Jiaxue Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Lin Xia
- School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Qian Wang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Xiaoyan Nie
- Department of Pharmacy Administration & Clinical Pharmacy, School of Pharmaceutical, Peking University, Beijing, China
| | - Yufei Feng
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Yi Fang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| |
Collapse
|
28
|
Cartaxo AL, Almeida J, Gualda EJ, Marsal M, Loza-Alvarez P, Brito C, Isidro IA. A computational diffusion model to study antibody transport within reconstructed tumor microenvironments. BMC Bioinformatics 2020; 21:529. [PMID: 33203360 PMCID: PMC7672975 DOI: 10.1186/s12859-020-03854-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022] Open
Abstract
Background Antibodies revolutionized cancer treatment over the past decades. Despite their successfully application, there are still challenges to overcome to improve efficacy, such as the heterogeneous distribution of antibodies within tumors. Tumor microenvironment features, such as the distribution of tumor and other cell types and the composition of the extracellular matrix may work together to hinder antibodies from reaching the target tumor cells. To understand these interactions, we propose a framework combining in vitro and in silico models. We took advantage of in vitro cancer models previously developed by our group, consisting of tumor cells and fibroblasts co-cultured in 3D within alginate capsules, for reconstruction of tumor microenvironment features.
Results In this work, an experimental-computational framework of antibody transport within alginate capsules was established, assuming a purely diffusive transport, combined with an exponential saturation effect that mimics the saturation of binding sites on the cell surface. Our tumor microenvironment in vitro models were challenged with a fluorescent antibody and its transport recorded using light sheet fluorescence microscopy. Diffusion and saturation parameters of the computational model were adjusted to reproduce the experimental antibody distribution, with root mean square error under 5%. This computational framework is flexible and can simulate different random distributions of tumor microenvironment elements (fibroblasts, cancer cells and collagen fibers) within the capsule. The random distribution algorithm can be tuned to follow the general patterns observed in the experimental models. Conclusions We present a computational and microscopy framework to track and simulate antibody transport within the tumor microenvironment that complements the previously established in vitro models platform. This framework paves the way to the development of a valuable tool to study the influence of different components of the tumor microenvironment on antibody transport.
Collapse
Affiliation(s)
- Ana Luísa Cartaxo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Jaime Almeida
- Departamento de Geologia, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal.,Instituto Dom Luiz, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Emilio J Gualda
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Maria Marsal
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Pablo Loza-Alvarez
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Inês A Isidro
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal. .,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
29
|
Luo MM, Usmani SZ, Mateos MV, Nahi H, Chari A, San-Miguel J, Touzeau C, Suzuki K, Kaiser M, Carson R, Heuck C, Qi M, Zhou H, Sun YN, Parasrampuria DA. Exposure-Response and Population Pharmacokinetic Analyses of a Novel Subcutaneous Formulation of Daratumumab Administered to Multiple Myeloma Patients. J Clin Pharmacol 2020; 61:614-627. [PMID: 33145788 PMCID: PMC8048619 DOI: 10.1002/jcph.1771] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 12/24/2022]
Abstract
We report the population pharmacokinetic (PK) and exposure-response analyses of a novel subcutaneous formulation of daratumumab (DARA) using data from 3 DARA subcutaneous monotherapy studies (PAVO Part 2, MMY1008, COLUMBA) and 1 combination therapy study (PLEIADES). Results were based on 5159 PK samples from 742 patients (DARA 1800 mg subcutaneously, n = 487 [monotherapy, n = 288; combination therapy, n = 199]; DARA 16 mg/kg intravenously, n = 255 [all monotherapy, in COLUMBA]; age, 33-92 years; weight, 28.6-147.6 kg). Subcutaneous and intravenous DARA monotherapies were administered once every week for cycles 1-2, once every 2 weeks for cycles 3-6, and once every 4 weeks thereafter (1 cycle is 28 days). The subcutaneous DARA combination therapy was administered with the adaptation of corresponding standard-of-care regimens. PK samples were collected between cycle 1 and cycle 12. Among monotherapy studies, throughout the treatment period, subcutaneous DARA provided similar/slightly higher trough concentrations (Ctrough ) versus intravenous DARA, with lower maximum concentrations and smaller peak-to-trough fluctuations. The PK profile was consistent between subcutaneous DARA monotherapy and combination therapies. The exposure-response relationship between daratumumab PK and efficacy or safety end points was similar for subcutaneous and intravenous DARA. Although the ≤65-kg subgroup reported a higher incidence of neutropenia, no relationship was found between the incidence of neutropenia and exposure, which was attributed, in part, to the preexisting imbalance in neutropenia between subcutaneous DARA (45.5%) and intravenous DARA (19%) in patients ≤50 kg. A flat relationship was observed between body weight and any grade and at least grade 3 infections. The results support the DARA 1800-mg subcutaneous flat dose as an alternative to the approved intravenous DARA 16 mg/kg.
Collapse
Affiliation(s)
- Man Melody Luo
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Saad Z Usmani
- Levine Cancer Institute/Atrium Health, Charlotte, North Carolina, USA
| | - Maria-Victoria Mateos
- University Hospital of Salamanca/IBSAL/Cancer Research Center-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Hareth Nahi
- Department of Medicine, Division of Hematology, Karolinska Institute, Karolinska University Hospital at Huddinge, Stockholm, Sweden
| | - Ajai Chari
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jesus San-Miguel
- Clínica Universidad de Navarra-CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | - Cyrille Touzeau
- Hematology Department, University Hospital Hôtel-Dieu, Nantes, France
| | - Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Martin Kaiser
- The Institute of Cancer Research and the Royal Marsden Hospital, London, UK
| | - Robin Carson
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Christoph Heuck
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Ming Qi
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Honghui Zhou
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Yu-Nien Sun
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | | |
Collapse
|
30
|
Mocenigo M, Porchetta A, Rossetti M, Brass E, Tonini L, Puzzi L, Tagliabue E, Triulzi T, Marini B, Ricci F, Ippodrino R. Rapid, Cost-Effective Peptide/Nucleic Acid-Based Platform for Therapeutic Antibody Monitoring in Clinical Samples. ACS Sens 2020; 5:3109-3115. [PMID: 32909731 DOI: 10.1021/acssensors.0c01046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We demonstrate here a homogeneous assay, named NanoHybrid, for monoclonal antibody quantification directly in serum samples in a single-step format. NanoHybrid is composed of both synthetic peptide nucleic acids (PNAs) and nucleic acid strands conjugated to recognition elements and optical labels and is designed to allow fast fluorescence quantification of a therapeutic antibody. More specifically, we have characterized our analytical assay for the detection of trastuzumab (Herceptin), a monoclonal antibody (mAb) drug used for breast cancer treatment and for tumors overexpressing the HER2/neu protein. We show here that NanoHybrid is capable of performing fast drug quantification directly in blood serum. The results obtained with a pool of samples from breast cancer patients under trastuzumab treatment are compared with CE-IVD ELISA (enzyme-linked immunosorbent assay) showing a good agreement (Cohen's K = 0.729). Due to the modular nature of the NanoHybrid platform, this technology can be programmed to potentially detect and quantify any antibody for which a high-affinity recognition element has been characterized. We envision the application of NanoHybrid in a point-of-care (POC) drug monitoring system based on disposable kits for therapeutic drug management.
Collapse
Affiliation(s)
- Marco Mocenigo
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
- Molecular Genetics and Biotechnology PhD Study Programme, University of Nova Gorica, Vipavska 13, 5000 Nova Gorica, Slovenia
| | - Alessandro Porchetta
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Marianna Rossetti
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Erik Brass
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Lucia Tonini
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Luca Puzzi
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Elda Tagliabue
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Tiziana Triulzi
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Bruna Marini
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| | - Francesco Ricci
- Department of Chemistry, University of Rome, Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Rudy Ippodrino
- Ulisse BioMed Labs, Area Science Park, SS 14, km 163.5, 34149 Trieste, Italy
| |
Collapse
|
31
|
Chang JH, Cheng CC, Lu YY, Chen YC, Chen SA, Chen YJ. Trastuzumab increases pulmonary vein arrhythmogenesis through modulating pulmonary vein electrical and conduction properties via phosphatidylinositol 3-kinase signaling. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:865-870. [PMID: 32774807 PMCID: PMC7395187 DOI: 10.22038/ijbms.2020.44651.10432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/09/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Drug-induced atrial fibrillation (AF) is considered an adverse effect of chemotherapeutic drugs. AF is a crucial risk factor for stroke, heart failure, myocardial infarction, and mortality. Pulmonary veins (PVs) are considered triggers inducing AF, and the sinoatrial node (SAN) may modulate PV activity and participate in AF genesis. AF was associated with early discontinuation of trastuzumab in patients with breast cancer. However, whether trastuzumab directly modulates the electrophysiological characteristics of PV and SAN remains unclear. MATERIALS AND METHODS ECG and conventional microelectrode system were used to record rabbit heart rhythm in vivo and electrical activities in vitro from isolated SAN, PV, and SAN-PV preparations. RESULTS Trastuzumab reduced the beating rate in isolated PV and SAN preparations at 1, 10, and 30 μM (particularly in isolated SAN preparations) and induced burst firings in isolated PV preparations at 10 μΜ. In addition, trastuzumab (10 μM) induced SAN-PV conduction block and burst firings, which were blocked by wortmannin (a PI3K inhibitor, 100 nM). Similarly, ECG recordings showed that acute intravenous administration of trastuzumab (10 mg/kg) reduced rabbit heart rates. CONCLUSION Trastuzumab increased PV arrhythmogenesis through interfering with PI3K signaling, which may contribute to the genesis of AF.
Collapse
Affiliation(s)
- Jun-Hei Chang
- Department of Medical, Tri-Service General Hospital Songshan Branch, National Defense Medical Center, Taipei, Taiwan
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Chen-Chuan Cheng
- Department of Cardiology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ann Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Jen Chen
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
32
|
Rossetti M, Brannetti S, Mocenigo M, Marini B, Ippodrino R, Porchetta A. Harnessing Effective Molarity to Design an Electrochemical DNA‐based Platform for Clinically Relevant Antibody Detection. Angew Chem Int Ed Engl 2020; 59:14973-14978. [DOI: 10.1002/anie.202005124] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Marianna Rossetti
- Department of Chemistry University of Rome Tor Vergata Via della Ricerca Scientifica 00133 Rome Italy
| | - Simone Brannetti
- Department of Chemistry University of Rome Tor Vergata Via della Ricerca Scientifica 00133 Rome Italy
| | - Marco Mocenigo
- Ulisse BioMed S.r.l. Area Science Park 34149 Trieste Italy
| | - Bruna Marini
- Ulisse BioMed S.r.l. Area Science Park 34149 Trieste Italy
| | - Rudy Ippodrino
- Ulisse BioMed S.r.l. Area Science Park 34149 Trieste Italy
| | - Alessandro Porchetta
- Department of Chemistry University of Rome Tor Vergata Via della Ricerca Scientifica 00133 Rome Italy
| |
Collapse
|
33
|
Rossetti M, Brannetti S, Mocenigo M, Marini B, Ippodrino R, Porchetta A. Harnessing Effective Molarity to Design an Electrochemical DNA‐based Platform for Clinically Relevant Antibody Detection. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Marianna Rossetti
- Department of Chemistry University of Rome Tor Vergata Via della Ricerca Scientifica 00133 Rome Italy
| | - Simone Brannetti
- Department of Chemistry University of Rome Tor Vergata Via della Ricerca Scientifica 00133 Rome Italy
| | - Marco Mocenigo
- Ulisse BioMed S.r.l. Area Science Park 34149 Trieste Italy
| | - Bruna Marini
- Ulisse BioMed S.r.l. Area Science Park 34149 Trieste Italy
| | - Rudy Ippodrino
- Ulisse BioMed S.r.l. Area Science Park 34149 Trieste Italy
| | - Alessandro Porchetta
- Department of Chemistry University of Rome Tor Vergata Via della Ricerca Scientifica 00133 Rome Italy
| |
Collapse
|
34
|
Ternant D, Azzopardi N, Raoul W, Bejan-Angoulvant T, Paintaud G. Influence of Antigen Mass on the Pharmacokinetics of Therapeutic Antibodies in Humans. Clin Pharmacokinet 2020; 58:169-187. [PMID: 29802542 DOI: 10.1007/s40262-018-0680-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic antibodies are increasingly used to treat various diseases, including neoplasms and chronic inflammatory diseases. Antibodies exhibit complex pharmacokinetic properties, notably owing to the influence of antigen mass, i.e. the amount of antigenic targets to which the monoclonal antibody binds specifically. This review focuses on the influence of antigen mass on the pharmacokinetics of therapeutic antibodies quantified by pharmacokinetic modelling in humans. Out of 159 pharmacokinetic studies, 85 reported an influence of antigen mass. This influence led to non-linear elimination decay in 50 publications, which was described using target-mediated drug disposition or derived models, as quasi-steady-state, irreversible binding and Michaelis-Menten models. In 35 publications, the pharmacokinetics was apparently linear and the influence of antigen mass was described as a covariate of pharmacokinetic parameters. If some reported covariates, such as the circulating antigen level or tumour size, are likely to be correlated to antigen mass, others, such as disease activity or disease type, may contain little information on the amount of antigenic targets. In some cases, antigen targets exist in different forms, notably in the circulation and expressed at the cell surface. The influence of antigen mass should be soundly described during the early clinical phases of drug development. To maximise therapeutic efficacy, sufficient antibody doses should be administered to ensure the saturation of antigen targets by therapeutic antibodies in all patients. If necessary, antigen mass should be taken into account in routine clinical practice.
Collapse
Affiliation(s)
- David Ternant
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France. .,Department of Medical Pharmacology, CHRU de Tours, Tours University Hospital, 2 boulevard Tonnellé, 37044, Tours Cedex, France.
| | | | - William Raoul
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France
| | - Theodora Bejan-Angoulvant
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours University Hospital, 2 boulevard Tonnellé, 37044, Tours Cedex, France
| | - Gilles Paintaud
- Université de Tours, EA7501 GICC, Team PATCH, Tours, France.,Department of Medical Pharmacology, CHRU de Tours, Tours University Hospital, 2 boulevard Tonnellé, 37044, Tours Cedex, France
| |
Collapse
|
35
|
Leemasawat K, Phrommintikul A, Chattipakorn SC, Chattipakorn N. Mechanisms and potential interventions associated with the cardiotoxicity of ErbB2-targeted drugs: Insights from in vitro, in vivo, and clinical studies in breast cancer patients. Cell Mol Life Sci 2020; 77:1571-1589. [PMID: 31650186 PMCID: PMC11104997 DOI: 10.1007/s00018-019-03340-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/22/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most frequently occurring cancer among women worldwide. Human epidermal growth factor receptor 2 (HER2 or ErbB2) is overexpressed in between 20 and 25% of invasive breast cancers and is associated with poor prognosis. Trastuzumab, an anti-ErbB2 monoclonal antibody, reduces cancer recurrence and mortality in HER2-positive breast cancer patients, but unexpectedly induces cardiac dysfunction, especially when used in combination with anthracycline-based chemotherapy. Novel approved ErbB2-targeting drugs, including lapatinib, pertuzumab, and trastuzumab-emtansine, also potentially cause cardiotoxicity, although early clinical studies demonstrate their cardiac safety profile. Unfortunately, the mechanism involved in causing the cardiotoxicity is still not completely understood. In addition, the use of preventive interventions against trastuzumab-induced cardiac dysfunction, including angiotensin-converting enzyme inhibitors and beta-blockers, remain controversial. Thus, this review aims to summarize and discuss the evidence currently available from in vitro, in vivo, and clinical studies regarding the mechanism and potential interventions associated with the cardiotoxicity of ErbB2-targeted drugs.
Collapse
Affiliation(s)
- Krit Leemasawat
- Division of Cardiovascular Diseases, Department of Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Arintaya Phrommintikul
- Division of Cardiovascular Diseases, Department of Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
36
|
González García J, Gutiérrez Nicolás F, Ramos Díaz R, Nazco Casariego GJ, Viña Romero MM, Llabres Martinez M, Llanos Muñoz M, Batista López JN, Jiménez Sosa A, Ceballos Lenza I, Cruz Jurado J. Pharmacokinetics of Trastuzumab After Subcutaneous and Intravenous Administration in Obese Patients. Ann Pharmacother 2020; 54:775-779. [PMID: 31971005 DOI: 10.1177/1060028020902318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Subcutaneous trastuzumab (T-SC) administration does not allow the historical target concentration of 20 µg/mL for efficacy to be reached, from the start of treatment in patients with a body mass index (BMI) >30 kg/m2. Objectives: To analyze the influence of the strategy of dosification (fixed vs adjusted patient's body weight dose) on the initial minimum plasma concentration (Cmin) of trastuzumab in obese patients. Methods: This was an observational, prospective study, which included patients with HER2-positive nonmetastatic breast cancer treated with trastuzumab. The determination of the Cmin of trastuzumab was performed on day +21 of the first cycle using the ELISA technique. Patients were stratified according to the strategy of dosification and BMI. Results: A total of 50 patients were included; 16 patients received the drug intravenously and 34 in a fixed dosage subcutaneous (T-SC) regimen. The proportion of patients who achieved an adequate plasma concentration since the beginning of treatment was significantly higher when the drug was administered intravenously (93.8% vs 67.6%, P = 0.042). These differences are especially greater in T-SC patients with BMI >30 kg/m2, with only 20% of patients exceeding the pharmacokinetic target. Conclusion and Relevance: Our study suggests that trastuzumab SC fixed dose of 600 mg is not equivalent to IV administration, especially in obese patients. An adequate trastuzumab exposure in this population needs patient weight-adjusted IV dosage in the first administration. The clinical relevance of these findings remains to be elucidated, and further research, including larger controlled trials, is warranted.
Collapse
Affiliation(s)
| | | | - Ruth Ramos Díaz
- Fundación Canaria para la Investigación Sanitaria (FUNCANIS), Santa Cruz de Tenerife, La Laguna, Spain
| | | | | | | | - Marta Llanos Muñoz
- Complejo Hospitalario Universitario de Canarias, Santa Cruz de Tenerife, La Laguna, Spain
| | | | - Alejandro Jiménez Sosa
- Complejo Hospitalario Universitario de Canarias, Santa Cruz de Tenerife, La Laguna, Spain
| | - Isaac Ceballos Lenza
- Complejo Hospitalario Universitario de Canarias, Santa Cruz de Tenerife, La Laguna, Spain
| | - Josefina Cruz Jurado
- Complejo Hospitalario Universitario de Canarias, Santa Cruz de Tenerife, La Laguna, Spain
| |
Collapse
|
37
|
|
38
|
Styles IK, Feeney OM, Nguyen TH, Brundel DHS, Kang DW, Clift R, McIntosh MP, Porter CJH. Removal of interstitial hyaluronan with recombinant human hyaluronidase improves the systemic and lymphatic uptake of cetuximab in rats. J Control Release 2019; 315:85-96. [PMID: 31655131 DOI: 10.1016/j.jconrel.2019.10.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 11/28/2022]
Abstract
Interstitial, e.g. subcutaneous (SC) or intradermal (ID), administration of monoclonal antibodies (mAb) is less invasive than intravenous administration and leads to mAb uptake into both lymphatic and blood capillaries draining the injection site. Interstitial administration, however, is hindered by the presence of hyaluronan (HA), a glycosaminoglycan that is a major fluid barrier in the interstitial space. The transient removal of HA with recombinant human hyaluronidase (rHuPH20) helps facilitate the interstitial administration of often high therapeutic doses of mAb in the clinic. rHuPH20's impact on the systemic pharmacokinetics of several mAbs has been previously described, however effects on route of absorption (lymph vs blood) are unknown. The current study has therefore explored the lymphatic transport and bioavailability of cetuximab and trastuzumab after SC and ID coadministration in the presence and absence of rHuPH20 in rats. After SC administration cetuximab absolute bioavailability increased from 67 % to 80 % in the presence of rHuPH20. Cetuximab recovery in the lymphatics also increased after SC (35.8 % to 49.4 %) and ID (26.7 % to 58.8 %) administration in the presence of rHuPH20. When the injection volume (and therefore dose) was increased 10-fold in the presence of rHuPH20 cetuximab plasma exposure increased approximately linearly (12- and 8.9-fold respectively after SC and ID administration), although the proportional contribution of cetuximab lymphatic transport reduced slightly (6.2-fold increase for both administration routes). In contrast, co-administration with rHuPH20 did not lead to increases in plasma exposure for trastuzumab after SC or ID administration, most likely reflecting the fact that the reported absolute bioavailability of trastuzumab (in the absence of rHuPH20) is high (∼77-99 %). However, lymphatic transport of trastuzumab did increase when coadministered ID with rHuPH20 in spite of the lack of change to overall bioavailability. The data suggest that co-administration with rHuPH20 is able to increase the volume of mAb that can be administered interstitially, and in some instances can increase the amount absorbed into both the blood and the lymph. In the current studies the ability of rHuPH20 to enhance interstitial bioavailability was higher for cetuximab where intrinsic interstitial bioavailability was low, when compared to trastuzumab where interstitial bioavailability was high.
Collapse
Affiliation(s)
- Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Tri-Hung Nguyen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Daniel H S Brundel
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - David W Kang
- Halozyme Therapeutics Inc, 11388 Sorrento Valley Rd, San Diego, California 92121, USA
| | - Renee Clift
- Halozyme Therapeutics Inc, 11388 Sorrento Valley Rd, San Diego, California 92121, USA
| | - Michelle P McIntosh
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.
| |
Collapse
|
39
|
Chung S, Nguyen V, Lin YL, Lafrance-Vanasse J, Scales SJ, Lin K, Deng R, Williams K, Sperinde G, Li JJ, Zheng K, Sukumaran S, Tesar D, Ernst JA, Fischer S, Lazar GA, Prabhu S, Song A. An in vitro FcRn- dependent transcytosis assay as a screening tool for predictive assessment of nonspecific clearance of antibody therapeutics in humans. MAbs 2019; 11:942-955. [PMID: 30982394 PMCID: PMC6601550 DOI: 10.1080/19420862.2019.1605270] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
A cell-based assay employing Madin–Darby canine kidney cells stably expressing human neonatal Fc receptor (FcRn) heavy chain and β2-microglobulin genes was developed to measure transcytosis of monoclonal antibodies (mAbs) under conditions relevant to the FcRn-mediated immunoglobulin G (IgG) salvage pathway. The FcRn-dependent transcytosis assay is modeled to reflect combined effects of nonspecific interactions between mAbs and cells, cellular uptake via pinocytosis, pH-dependent interactions with FcRn, and dynamics of intracellular trafficking and sorting mechanisms. Evaluation of 53 mAbs, including 30 marketed mAb drugs, revealed a notable correlation between the transcytosis readouts and clearance in humans. FcRn was required to promote efficient transcytosis of mAbs and contributed directly to the observed correlation. Furthermore, the transcytosis assay correctly predicted rank order of clearance of glycosylation and Fv charge variants of Fc-containing proteins. These results strongly support the utility of this assay as a cost-effective and animal-sparing screening tool for evaluation of mAb-based drug candidates during lead selection, optimization, and process development for desired pharmacokinetic properties.
Collapse
Affiliation(s)
- Shan Chung
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Van Nguyen
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Yuwen Linda Lin
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | | | - Suzie J Scales
- c Department of Molecular Biology , Genentech Inc ., South San Francisco , CA , USA
| | - Kevin Lin
- d Department of Analytical Operations , Genentech Inc ., South San Francisco , CA , USA
| | - Rong Deng
- e Department of Clinical Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kathi Williams
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Gizette Sperinde
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Juan Jenny Li
- f Department of Biochemistry and Cellular Pharmacology , Genentech Inc ., South San Francisco , CA , USA
| | - Kai Zheng
- g Department of Late Stage Pharmaceutical Development , Genentech Inc ., South San Francisco , CA , USA
| | - Siddharth Sukumaran
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - Devin Tesar
- i Department of Drug Delivery , Genentech Inc ., South San Francisco , CA , USA
| | - James A Ernst
- b Department of Protein Chemistry , Genentech Inc ., South San Francisco , CA , USA
| | - Saloumeh Fischer
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| | - Greg A Lazar
- j Department of Antibody Engineering , Genentech Inc ., South San Francisco , CA , USA
| | - Saileta Prabhu
- h Department of Pharmacokinetics & Pharmacodynamics , Genentech Inc ., South San Francisco , CA , USA
| | - An Song
- a Department of BioAnalytical Sciences , Genentech Inc ., South San Francisco , CA , USA
| |
Collapse
|
40
|
Pharmacokinetic and exploratory exposure-response analysis of pertuzumab in patients with operable HER2-positive early breast cancer in the APHINITY study. Cancer Chemother Pharmacol 2019; 83:1147-1158. [PMID: 30976844 PMCID: PMC6499763 DOI: 10.1007/s00280-019-03826-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/25/2019] [Indexed: 12/22/2022]
Abstract
Purpose To characterize the pharmacokinetics (PK) of, and perform an exploratory exposure–response (E–R) analysis for, pertuzumab in patients with HER2-positive early breast cancer (EBC) within the APHINITY study (NCT01358877, BIG 4–11/BO25126/TOC4939G). Methods A previously developed pertuzumab two-compartment linear population pharmacokinetic (popPK) model was subjected to external validation to examine appropriateness for describing pertuzumab concentrations from the APHINITY study. Pharmacokinetic drug–drug interactions (DDIs) between pertuzumab, trastuzumab, and chemotherapy were assessed by comparing observed serum or plasma Cmax, Cmin, and AUClast geometric mean ratios with 90% CIs. Predictions of pertuzumab Cmax,ss, Cmin,ss, and AUCss were derived from individual parameter estimates and used in an exploratory E–R analysis. Results Using data from 72 patients, based on goodness-of-fit, the popPK model was deemed appropriate for predictions of individual exposures for subsequent comparisons to historical data, assessment of DDIs, and E–R analyses. No evidence of DDIs for pertuzumab on trastuzumab, trastuzumab on pertuzumab, or pertuzumab on chemotherapy PK was observed. Analyses of differences in exposure between patients with and without invasive disease-free survival events did not indicate improved efficacy with increased exposure. Overall Grade ≥ 3 diarrhea prevalence was higher with pertuzumab versus placebo, but was not greater with increasing pertuzumab exposure. No apparent E–R relationship was suggested with respect to other grade ≥ 3 AEs. Conclusion Overall, the limited available data from this exploratory study suggest that no dose adjustments are needed for pertuzumab when administered in combination with trastuzumab and an EBC chemotherapy regimen. Electronic supplementary material The online version of this article (10.1007/s00280-019-03826-1) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Kassem L, Shohdy KS, Abdel-Azeez AM, Attia H. Is the Fixed-Dose Intravenous Trastuzumab Policy Warranted in Limited-Resource Settings? J Glob Oncol 2019; 5:1-3. [PMID: 30676841 PMCID: PMC6426478 DOI: 10.1200/jgo.18.00155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Loay Kassem
- Cairo University Kasr Alainy Faculty of Medicine, Cairo, Egypt
| | | | | | - Hanaa Attia
- Cairo University Kasr Alainy Faculty of Medicine, Cairo, Egypt
| |
Collapse
|
42
|
Chandra F, Zaks L, Zhu A. Survival Prolongation Index as a Novel Metric to Assess Anti-Tumor Activity in Xenograft Models. AAPS JOURNAL 2019; 21:16. [PMID: 30627814 DOI: 10.1208/s12248-018-0284-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
A single efficacy metric quantifying anti-tumor activity in xenograft models is useful in evaluating different tumors' drug sensitivity and dose-response of an anti-tumor agent. Commonly used metrics include the ratio of tumor volume in treated vs. control mice (T/C), tumor growth inhibition (TGI), ratio of area under the curve (AUC), and growth rate inhibition (GRI). However, these metrics have some limitations. In particular, for biologics with long half-lives, tumor volume (TV) of treated xenografts displays a delay in volume reduction (and in some cases, complete regression) followed by a growth rebound. These observed data cannot be described by exponential functions, which is the underlying assumption of TGI and GRI, and the fit depends on how long the tumor volumes are monitored. On the other hand, T/C and TGI only utilizes information from one chosen time point. Here, we propose a new metric called Survival Prolongation Index (SPI), calculated as the time for drug-treated TV to reach a certain size (e.g., 600 mm3) divided by the time for control TV to reach 600mm3 and therefore not dependent on the chosen final time point tf. Simulations were conducted under different scenarios (i.e., exponential vs. saturable growth, linear vs. nonlinear kill function). For all cases, SPI is the most linear and growth-rate independent metric. Subsequently, a literature analysis was conducted using 11 drugs to evaluate the correlation between pre-clinically obtained SPI and clinical overall response. This retrospective analysis of approved drugs suggests that a predicted SPI of 2 is necessary for clinical response.
Collapse
Affiliation(s)
- Fiona Chandra
- Translation Modeling and Simulation, DMPK, Takeda Pharmaceuticals, 35 Landsdowne St, Cambridge, Massachusetts, 02139, USA.
| | - Lihi Zaks
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andy Zhu
- Translation Modeling and Simulation, DMPK, Takeda Pharmaceuticals, 35 Landsdowne St, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
43
|
Cheng Y, Wu L, Liu X, Zhao Y, Liu C, Chen Q, Sun T, Zheng Q. Population pharmacokinetics and individualized lobaplatin regimen for the treatment of Chinese small cell lung cancer in the elderly. Medicine (Baltimore) 2019; 98:e14136. [PMID: 30653145 PMCID: PMC6370119 DOI: 10.1097/md.0000000000014136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Lobaplatin (LBP) is a third-generation platinum compound. MATERIAL AND METHODS This prospective study was performed in 7 institutions in 2014-2016. Elderly small cell lung cancer (SCLC) patients (≥65 years old) were divided into 2 groups to receive LBP regimens according to endogenous creatinine clearance rate (Ccr). LBP was administered at 30 and 20 mg/m in groups A (Ccr ≥ 80 ml/min) and B (60 ml/min ≤ Ccr < 80 ml/min), respectively. The primary endpoint was plasma LBP concentrations. Secondary endpoints were safety and efficacy parameters, including progression-free survival (PFS) and overall survival (OS). RESULTS One-hundred patients were enrolled. Median PFS and OS in groups A and B were 155 vs170 days and 306 vs 272 days, respectively. The rates of grade III/IV AEs in groups A and B were 60.8% (n = 31) and 51.0% (n = 25), respectively. In population pharmacokinetics, the area under the curve (AUC) value for group B was 39% lower than that of group A. With LBP administration based on body surface area (BSA), AUC differences between individuals were small. CONCLUSION With Ccr ≥ 60 ml/min, BSA based administration is necessary. Meanwhile, LBP-based regimens are reliable in treating elderly patients with SCLC.
Collapse
Affiliation(s)
| | - Lin Wu
- Hunan Cancer Hospital, Changsha
| | | | | | - Chunling Liu
- Affiliated Cancer Hospital of Xinjiang Medical University, Urumchi
| | - Qun Chen
- Fuzhou Pulmonary Hospital of Fujian, Fuzhou
| | - Tao Sun
- Liaoning Cancer Hospital, Shenyang
| | - Qingshan Zheng
- Clinical Research Centre of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
Lucas AT, Robinson R, Schorzman AN, Piscitelli JA, Razo JF, Zamboni WC. Pharmacologic Considerations in the Disposition of Antibodies and Antibody-Drug Conjugates in Preclinical Models and in Patients. Antibodies (Basel) 2019; 8:E3. [PMID: 31544809 PMCID: PMC6640706 DOI: 10.3390/antib8010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 12/11/2022] Open
Abstract
The rapid advancement in the development of therapeutic proteins, including monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), has created a novel mechanism to selectively deliver highly potent cytotoxic agents in the treatment of cancer. These agents provide numerous benefits compared to traditional small molecule drugs, though their clinical use still requires optimization. The pharmacology of mAbs/ADCs is complex and because ADCs are comprised of multiple components, individual agent characteristics and patient variables can affect their disposition. To further improve the clinical use and rational development of these agents, it is imperative to comprehend the complex mechanisms employed by antibody-based agents in traversing numerous biological barriers and how agent/patient factors affect tumor delivery, toxicities, efficacy, and ultimately, biodistribution. This review provides an updated summary of factors known to affect the disposition of mAbs/ADCs in development and in clinical use, as well as how these factors should be considered in the selection and design of preclinical studies of ADC agents in development.
Collapse
Affiliation(s)
- Andrew T Lucas
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Ryan Robinson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Joseph A Piscitelli
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - Juan F Razo
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- University of North Carolina (UNC), Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
45
|
Kirschbrown WP, Wynne C, Kågedal M, Wada R, Li H, Wang B, Nijem I, Badovinac Crnjevic T, Gasser H, Heeson S, Eng-Wong J, Garg A. Development of a Subcutaneous Fixed-Dose Combination of Pertuzumab and Trastuzumab: Results From the Phase Ib Dose-Finding Study. J Clin Pharmacol 2018; 59:702-716. [PMID: 30570763 PMCID: PMC7027517 DOI: 10.1002/jcph.1362] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/26/2018] [Indexed: 12/18/2022]
Abstract
Adding pertuzumab to trastuzumab (both monoclonal antibodies targeting human epidermal growth factor receptor 2 [HER2]) has proven survival benefits when combined with chemotherapy for patients with HER2-positive breast cancer. The combination of pertuzumab and trastuzumab together in 1 vial for subcutaneous (SC) administration is being developed as a ready-to-use formulation to reduce the treatment burden on patients while improving healthcare efficiency. An open-label, 2-part, phase Ib dose-finding study (NCT02738970) was undertaken in healthy male volunteers (part 1) and female patients with HER2-postive early breast cancer who had completed standard (neo)adjuvant treatment (part 2). This study aimed to identify an SC pertuzumab dose given with recombinant human hyaluronidase that results in comparable exposure to that of the intravenous (IV) pertuzumab dose, based on pertuzumab serum trough concentration and area under the serum concentration-time curve. Pharmacokinetics (PK), safety, and tolerability of a single dose of SC pertuzumab given alone or in a fixed-dose combination (comixed or coformulated) with trastuzumab were also assessed. A maintenance dose of 600 mg for SC pertuzumab resulted in an equivalent exposure to that of IV pertuzumab, and no new safety signals were identified for SC pertuzumab or trastuzumab. A loading dose of 1200 mg for SC pertuzumab was selected based on approximate dose proportionality. The PK and safety results support further development of a fixed-dose coformulation combination of pertuzumab and trastuzumab for SC administration, which will be investigated in an upcoming phase III trial in patients with HER2-positive early breast cancer.
Collapse
Affiliation(s)
| | - Chris Wynne
- Christchurch Clinical Studies Trust, Christchurch, New Zealand
| | | | | | | | - Bei Wang
- Genentech, Inc., South San Francisco, CA, USA
| | - Ihsan Nijem
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | - Amit Garg
- Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
46
|
Inotai A, Ágh T, Karpenko AW, Zemplényi A, Kaló Z. Behind the subcutaneous trastuzumab hype: evaluation of benefits and their transferability to Central Eastern European countries. Expert Rev Pharmacoecon Outcomes Res 2018; 19:105-113. [DOI: 10.1080/14737167.2019.1554437] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- András Inotai
- Syreon Research Institute, Budapest, Hungary
- Department of Health Policy and Health Economics, Eötvös Loránd University (ELTE), Budapest, Hungary
| | - Tamás Ágh
- Syreon Research Institute, Budapest, Hungary
| | - Alexei Willem Karpenko
- Erasmus School of Health Policy & Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Antal Zemplényi
- Syreon Research Institute, Budapest, Hungary
- Department of Pharmacoeconomics, University of Pécs, Pécs, Hungary
| | - Zoltán Kaló
- Syreon Research Institute, Budapest, Hungary
- Department of Health Policy and Health Economics, Eötvös Loránd University (ELTE), Budapest, Hungary
| |
Collapse
|
47
|
Quartino AL, Li H, Kirschbrown WP, Mangat R, Wada DR, Garg A, Jin JY, Lum B. Population pharmacokinetic and covariate analyses of intravenous trastuzumab (Herceptin ®), a HER2-targeted monoclonal antibody, in patients with a variety of solid tumors. Cancer Chemother Pharmacol 2018; 83:329-340. [PMID: 30467591 PMCID: PMC6394489 DOI: 10.1007/s00280-018-3728-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE The aim of the study was to characterize the population pharmacokinetics (PK) of the intravenous formulation of trastuzumab, assess the impact of patient and pathological covariates on trastuzumab PK, and perform simulations to support dosing recommendations in special situations. METHODS Serum trastuzumab concentrations were obtained from 1582 patients with metastatic breast cancer (MBC), early breast cancer (EBC), advanced gastric cancer (AGC), or other tumor types/healthy volunteers in 18 phase I, II, and III trials and analyzed by nonlinear mixed-effects modeling. RESULTS A two-compartment model with parallel linear and nonlinear elimination best described the data. During treatment, linear clearance (CL) dominated, resulting in a total CL of 0.173-0.337 L/day, which is similar to other IgG1 monoclonal antibodies. Covariates influencing CL were baseline body weight, aspartate aminotransferase, albumin, gastric cancer, and the presence of liver metastases. MBC and EBC had similar PK parameters, while CL was higher in AGC. Simulations indicated that at least 95% of patients with BC reach concentrations < 1 µg/mL (~ 97% washout) by 7 months. A dose delay in BC or AGC patients of > 1 week would take approximately 6 weeks to get back within steady-state exposure range. CONCLUSIONS Trastuzumab PK for the intravenous formulation was well-described across cancer types, disease status, and regimens. No dose adjustment is required for any of the identified patient covariates. A 7-month serum washout period for trastuzumab is recommended. A reloading dose is required if a maintenance dose is missed by > 1 week.
Collapse
Affiliation(s)
- Angelica L Quartino
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA.
| | - Hanbin Li
- Certara, L.P., 845 Oak Grove Ave, Menlo Park, CA, 94025, USA
| | | | - Ranvir Mangat
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA.,Insight Rx, 233 Stanyan Street, San Francisco, CA, 94118, USA
| | - D Russell Wada
- Certara, L.P., 845 Oak Grove Ave, Menlo Park, CA, 94025, USA
| | - Amit Garg
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA
| | - Jin Y Jin
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA
| | - Bert Lum
- Genentech, Inc., 1 DNA Way, Mail Stop 463A, South San Francisco, CA, 94080, USA
| |
Collapse
|
48
|
Jung KH, Ataseven B, Verrill M, Pivot X, De Laurentiis M, Al-Sakaff N, Lauer S, Shing M, Gligorov J, Azim HA. Adjuvant Subcutaneous Trastuzumab for HER2-Positive Early Breast Cancer: Subgroup Analyses of Safety and Active Medical Conditions by Body Weight in the SafeHer Phase III Study. Oncologist 2018; 23:1137-1143. [PMID: 30018134 PMCID: PMC6263135 DOI: 10.1634/theoncologist.2018-0065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/23/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND This SafeHer subgroup analysis assessed the safety of fixed-dose subcutaneous trastuzumab (H SC) as an adjuvant therapy in HER2-positive early breast cancer (EBC) by body weight. PATIENTS AND METHODS Patients with HER2-positive EBC not previously treated with anti-HER2 therapy received H SC 600 mg (every 3 weeks for 18 cycles), with neoadjuvant or adjuvant chemotherapy or without adjuvant chemotherapy. Adverse events (AEs) were assessed throughout treatment and at final follow-up (28 ±5 days after last treatment). Subgroups were categorized by body weight, Asian origin, and chemotherapy administration. All analyses were descriptive. RESULTS Of 2,577 patients enrolled, 2,573 received ≥1 dose of study medication and were included in this safety analysis. Median body weight at baseline was 67.0 kg (range 33.6-150.0 kg). Any-grade AEs occurred in 88.7% (2,282/2,573) of the overall population, versus 87.1% (590/677) of the lowest bodyweight quartile (≤59 kg), 90.0% (561/623) of the highest quartile (>77 kg), and 86.5% (327/378) of the Asian population. Grade ≥3 AEs occurred in 23.2% (596/2,573) of the overall population, 17.9% (121/677) of the lowest bodyweight quartile, 26.8% (167/623) of the highest quartile, and 15.3% (58/378) of the Asian population. The highest bodyweight quartile had the highest incidence of medical conditions at baseline (highest quartile, 75.6%; lowest quartile, 56.1%). CONCLUSION These data support the use of fixed-dose H SC as an adjuvant therapy in HER2-positive EBC and confirm the comparable safety profile of H SC in patients with low body weight or of Asian origin versus the overall population in SafeHer. ClinicalTrials.gov: NCT01566721. IMPLICATIONS FOR PRACTICE The safety profile of fixed-dose subcutaneous trastuzumab (H SC) was comparable between patients in the lowest bodyweight subgroup and the overall patient population, and also between patients of Asian origin (of whom a higher proportion often fall within the lower bodyweight quartiles) and the overall population. The safety data from this SafeHer subgroup analysis therefore support the use of fixed-dose H SC 600 mg administered every 3 weeks as an adjuvant therapy for patients with HER2-positive early breast cancer across different bodyweight subgroups and in the Asian patient population.
Collapse
Affiliation(s)
- Kyung Hae Jung
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Beyhan Ataseven
- Kliniken Essen-Mitte, Essen, Germany
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Mark Verrill
- Northern Centre for Cancer Care, Newcastle upon Tyne, UK
| | - Xavier Pivot
- Centre Paul Strauss, I'Institut Régional du Cancer, Strasbourg, France
| | | | | | | | - Mona Shing
- Genentech, Inc., South San Francisco, California, USA
| | | | - Hamdy A Azim
- Oncology Department, School of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
49
|
Lucas AT, Price LSL, Schorzman AN, Storrie M, Piscitelli JA, Razo J, Zamboni WC. Factors Affecting the Pharmacology of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:E10. [PMID: 31544862 PMCID: PMC6698819 DOI: 10.3390/antib7010010] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Major advances in therapeutic proteins, including antibody-drug conjugates (ADCs), have created revolutionary drug delivery systems in cancer over the past decade. While these immunoconjugate agents provide several advantages compared to their small-molecule counterparts, their clinical use is still in its infancy. The considerations in their development and clinical use are complex, and consist of multiple components and variables that can affect the pharmacologic characteristics. It is critical to understand the mechanisms employed by ADCs in navigating biological barriers and how these factors affect their biodistribution, delivery to tumors, efficacy, and toxicity. Thus, future studies are warranted to better understand the complex pharmacology and interaction between ADC carriers and biological systems, such as the mononuclear phagocyte system (MPS) and tumor microenvironment. This review provides an overview of factors that affect the pharmacologic profiles of ADC therapies that are currently in clinical use and development.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Lauren S L Price
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Mallory Storrie
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | | | - Juan Razo
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
50
|
A short review of the pharmacokinetic behavior of biological medicinal agents for the clinical practice. Microchem J 2018. [DOI: 10.1016/j.microc.2017.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|