1
|
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
|
2
|
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
|
3
|
A Physiologically Based Pharmacokinetic Framework for Quantifying Antibody Distribution Gradients from Tumors to Tumor-Draining Lymph Nodes. Antibodies (Basel) 2022; 11:antib11020028. [PMID: 35466281 PMCID: PMC9036243 DOI: 10.3390/antib11020028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/02/2022] Open
Abstract
Immune checkpoint blockades prescribed in the neoadjuvant setting are now under active investigation for many types of tumors, and many have shown early success. The primary tumor (PT) and tumor-draining lymph node (TDLN) immune factors, along with adequate therapeutic antibody distributions to the PT and TDLN, are critical for optimal immune activation and anti-tumor efficacy in neoadjuvant immunotherapy. However, it remains largely unknown how much of the antibody can be distributed into the PT-TDLN axis at different clinical scenarios. The goal of the current work is to build a physiologically based pharmacokinetic (PBPK) model framework capable of characterizing antibody distribution gradients in the PT-TDLN axis across various clinical and pathophysiological scenarios. The model was calibrated using clinical data from immuno-PET antibody-imaging studies quantifying antibody pharmacokinetics (PK) in the blood, PTs, and TDLNs. The effects of metastatic lesion location, tumor-induced compression, and inflammation, as well as surgery, on antibody concentration gradients in the PT-TDLN axis were characterized. The PBPK model serves as a valuable tool to predict antibody exposures in various types of tumors, metastases, and the associated lymph node, supporting effective immunotherapy.
Collapse
|
4
|
Campone M, Bachelot T, Treilleux I, Pistilli B, Salleron J, Seegers V, Arnedos M, Loussouarn D, Wang Q, Vanlemmens L, Jimenez M, Rios M, Diéras V, Leroux A, Paintaud G, Rezai K, André F, Lion M, Merlin JL. A phase II randomised study of preoperative trastuzumab alone or combined with everolimus in patients with early HER2-positive breast cancer and predictive biomarkers (RADHER trial). Eur J Cancer 2021; 158:169-180. [PMID: 34678678 DOI: 10.1016/j.ejca.2021.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/06/2021] [Accepted: 09/11/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Resistance to trastuzumab in breast cancer is an ongoing challenge. Clinical and biological effects of co-targeting HER2 and mammalian target of rapamycin (mTOR) in patients with HER2-positive early operable breast cancer via the addition of everolimus to preoperative trastuzumab were evaluated in a phase II randomised study. METHODS Patients were randomised 1:1 to receive trastuzumab (4 mg/kg initial dose then 2 mg/kg weekly for 5 weeks) alone or combined with everolimus (10 mg/day for 6 weeks) and then underwent surgery. Tumours were assessed by clinical examination and echography at the baseline and on treatment. The primary end-point was the clinical response rate at 6 weeks. Pathological response and safety were also evaluated. Baseline and surgery tumour samples were assessed by immunohistochemistry and multiplex immunoanalysis for predictive downstream effectors of the PI3K/AKT/mTOR and MAP kinase (MAPK) pathways. RESULTS Eighty-two patients were enrolled, 41 per arm. The clinical response rates were 34.1% and 43.9% with trastuzumab alone and combined with everolimus, respectively. Pathological response rates were 43.6% and 47.5%, respectively. Addition of everolimus increased toxicity, notably mucositis (82.5% versus 5.0%) and rash (57.5% versus 10.0%), but grade III/IV events were rare. No correlation between response to treatments and baseline candidate biomarkers was identified, except for PIK3CA mutations which were found to predict trastuzumab resistance. Significant changes were seen in several MAPK pathway effectors after combination therapy. CONCLUSIONS The addition of everolimus did not improve the efficacy, but induced MAPK signalling. Combination therapy to overcome pathway cross-talk should be considered to maximise the effectiveness of trastuzumab in this setting. ClinicalTrial.gov Identifier NCT00674414.
Collapse
Affiliation(s)
- Mario Campone
- Department of Medical Oncology/ Cancer Research Center UMR-INSERM U892/CNRS 6299/ Bioinformatics Unit, Institut de Cancérologie de L'Ouest, Nantes, France
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, INSERM U1052, Lyon, France
| | - Isabelle Treilleux
- Department of Pathology and Biopathology, Centre Léon Bérard, Lyon, France
| | | | - Julia Salleron
- Methodology and Biostatistics Unit, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, France
| | - Valérie Seegers
- Oncology Data Factory and Analytics, Institut de Cancérologie de L'Ouest, Nantes, France
| | - Monica Arnedos
- Department of Medicine, Gustave Roussy, Villejuif, France
| | | | - Qing Wang
- Genomic Platform-Cancer Research Center of Lyon, Centre Léon Bérard, Lyon, France
| | | | | | - Maria Rios
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-les-Nancy, France
| | | | - Agnès Leroux
- Department of Biopathology, Institut de Cancérologie de Lorraine, Université de Lorraine, CNRS UMR 7039, CRAN, Vandoeuvre-Les-Nancy, France
| | - Gilles Paintaud
- François Rabelais University, CNRS, UMR 7292, Genetics, Immunotherapy, Chemistry and Cancer, Tours, France
| | - Keyvan Rezai
- Radio-Pharmacology Department, Institut Curie-Hôpital Rene Huguenin, Saint-Cloud, France
| | - Fabrice André
- Department of Medicine, Gustave Roussy, Villejuif, France
| | - Maëva Lion
- Department of Biopathology, Institut de Cancérologie de Lorraine, Université de Lorraine, CNRS UMR 7039, CRAN, Vandoeuvre-Les-Nancy, France
| | - Jean-Louis Merlin
- Department of Biopathology, Institut de Cancérologie de Lorraine, Université de Lorraine, CNRS UMR 7039, CRAN, Vandoeuvre-Les-Nancy, France.
| |
Collapse
|
5
|
Ramos MK, Mandikian D, Sermeño LN, King A, Dent AT, Ho J, Ulufatu S, Lombana TN, Spiess C, Go MAT, Yu SF, Kamath AV, Ferl GZ, Boswell CA. Valency of HER2 targeting antibodies influences tumor cell internalization and penetration. Mol Cancer Ther 2021; 20:1956-1965. [PMID: 34253591 DOI: 10.1158/1535-7163.mct-20-1097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/19/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022]
Abstract
T cell Dependent Bispecific antibodies (TDBs) have been a major advancement in the the treatment of cancer that allow for improved targeting and efficacy for large molecule therapeutics. TDBs are comprised of one arm targeting a surface antigen on a cancer cell and another targeting an engaging surface antigen on a cytotoxic T cell. In order to impart this function, the antibody must be in a bispecific format as opposed to the more conventional bivalent format. Through in vitro and in vivo studies, we sought to determine the impact of changing antibody valency on solid tumor distribution and catabolism. A bivalent anti-HER2 antibody exhibited higher catabolism than its full-length monovalent binding counterpart in vivo by both invasive tissue harvesting and non-invasive SPECT-CT imaging despite similar systemic exposures for the two molecules. In order to determine what molecular factors drove in vivo distribution and uptake, we developed a mechanistic model for binding and catabolism of monovalent and bivalent HER2 antibodies in KPL4 cells. This model suggests that observed differences in cellular uptake of monovalent and bivalent antibodies are caused by the change in apparent affinity conferred by avidity as well as differences in internalization and degradation rates of receptor bound antibodies. To our knowledge, this is the first study to directly compare the targeting abilities of monovalent and bivalent full-length antibodies. These findings may inform diverse antibody therapeutic modalities including T cell redirecting therapies and drug delivery strategies relying upon receptor internalization.
Collapse
Affiliation(s)
| | | | | | - Anna King
- Genentech Research and Early Development, Genentech, Inc
| | - Alecia T Dent
- Genentech Research and Early Development, Genentech, Inc
| | | | | | | | | | | | | | | | - Gregory Z Ferl
- Genentech Research and Early Development, Genentech, Inc
| | | |
Collapse
|
6
|
Population pharmacokinetics and exposure-response relationship of trastuzumab and bevacizumab in early-stage breast cancer. Eur J Clin Pharmacol 2021; 77:1861-1873. [PMID: 34245336 DOI: 10.1007/s00228-021-03179-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 06/21/2021] [Indexed: 10/20/2022]
Abstract
AIMS To describe the sources of interindividual variability of bevacizumab and trastuzumab pharmacokinetics in early-stage breast cancer, and to study the relationship between exposure and both early clinical response and specific adverse events. PATIENTS AND METHODS Patients (n = 86) received 6 cycles of docetaxel + trastuzumab. Early tumour response was assessed by determination of the maximum standard uptake value (SUVmax) variation (ΔSUVmax) after 1 cycle using [18F]-fluorodeoxyglucose (FDG) PET. Early poor responders (ΔSUVmax < 70%) also received bevacizumab from cycle 3 to cycle 6. Sources of interindividual variability in pharmacokinetics of both antibodies were studied by population compartment modelling. Exposure as assessed by area under the concentration-versus-time curve (AUC) was compared between responders and non-responders and between patients experiencing specific adverse events or not. RESULTS A two-compartment model described the pharmacokinetics of both antibodies satisfactorily. Their central volume of distributions (Vc) increased with body surface area and their elimination half-lives were shorter (~14 days) than previously reported (~26-28 days). There was a time-dependent increase in trastuzumab Vc, positively correlated to baseline SUVmax. Bevacizumab elimination rate (k10) was positively correlated with ΔSUVmax measured at the end of the first cycle. Bevacizumab had no significantly influence on trastuzumab pharmacokinetics. No relationship between exposure and clinical response or occurrence of adverse events was found. CONCLUSION Tumour uptake as assessed by SUVmax influences the pharmacokinetics of bevacizumab and trastuzumab. In early-stage breast cancer, elimination half-lives of these therapeutic monoclonal antibodies may be shorter than those previously reported in more advanced disease.
Collapse
|
7
|
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
|
8
|
Modeling Pharmacokinetics and Pharmacodynamics of Therapeutic Antibodies: Progress, Challenges, and Future Directions. Pharmaceutics 2021; 13:pharmaceutics13030422. [PMID: 33800976 PMCID: PMC8003994 DOI: 10.3390/pharmaceutics13030422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022] Open
Abstract
With more than 90 approved drugs by 2020, therapeutic antibodies have played a central role in shifting the treatment landscape of many diseases, including autoimmune disorders and cancers. While showing many therapeutic advantages such as long half-life and highly selective actions, therapeutic antibodies still face many outstanding issues associated with their pharmacokinetics (PK) and pharmacodynamics (PD), including high variabilities, low tissue distributions, poorly-defined PK/PD characteristics for novel antibody formats, and high rates of treatment resistance. We have witnessed many successful cases applying PK/PD modeling to answer critical questions in therapeutic antibodies’ development and regulations. These models have yielded substantial insights into antibody PK/PD properties. This review summarized the progress, challenges, and future directions in modeling antibody PK/PD and highlighted the potential of applying mechanistic models addressing the development questions.
Collapse
|
9
|
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
|
10
|
Bensalem A, Ternant D. Pharmacokinetic Variability of Therapeutic Antibodies in Humans: A Comprehensive Review of Population Pharmacokinetic Modeling Publications. Clin Pharmacokinet 2020; 59:857-874. [DOI: 10.1007/s40262-020-00874-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
11
|
Padmanabhan R, Kheraldine HS, Meskin N, Vranic S, Al Moustafa AE. Crosstalk between HER2 and PD-1/PD-L1 in Breast Cancer: From Clinical Applications to Mathematical Models. Cancers (Basel) 2020; 12:E636. [PMID: 32164163 PMCID: PMC7139939 DOI: 10.3390/cancers12030636] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is one of the major causes of mortality in women worldwide. The most aggressive breast cancer subtypes are human epidermal growth factor receptor-positive (HER2+) and triple-negative breast cancers. Therapies targeting HER2 receptors have significantly improved HER2+ breast cancer patient outcomes. However, several recent studies have pointed out the deficiency of existing treatment protocols in combatting disease relapse and improving response rates to treatment. Overriding the inherent actions of the immune system to detect and annihilate cancer via the immune checkpoint pathways is one of the important hallmarks of cancer. Thus, restoration of these pathways by various means of immunomodulation has shown beneficial effects in the management of various types of cancers, including breast. We herein review the recent progress in the management of HER2+ breast cancer via HER2-targeted therapies, and its association with the programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) axis. In order to link research in the areas of medicine and mathematics and point out specific opportunities for providing efficient theoretical analysis related to HER2+ breast cancer management, we also review mathematical models pertaining to the dynamics of HER2+ breast cancer and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Regina Padmanabhan
- Department of Electrical Engineering, Qatar University, 2713 Doha, Qatar;
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
| | - Hadeel Shafeeq Kheraldine
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
- College of Pharmacy, QU Health, Qatar University, 2713 Doha, Qatar
| | - Nader Meskin
- Department of Electrical Engineering, Qatar University, 2713 Doha, Qatar;
| | - Semir Vranic
- College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
| | - Ala-Eddin Al Moustafa
- Biomedical Research Centre, Qatar University, 2713 Doha, Qatar;
- College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
| |
Collapse
|
12
|
Population Pharmacokinetics of MCLA-128, a HER2/HER3 Bispecific Monoclonal Antibody, in Patients with Solid Tumors. Clin Pharmacokinet 2020; 59:875-884. [PMID: 32006223 DOI: 10.1007/s40262-020-00858-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES MCLA-128 is a bispecific monoclonal antibody targeting the HER2 and HER3 receptors and is in development to overcome HER3-mediated resistance to anti-HER2 therapies. The aims of this analysis were to characterize the population pharmacokinetics of MCLA-128 in patients with various solid tumors, to evaluate patient-related factors that affect the disposition of MCLA-128, and to assess whether flat dosing is appropriate. METHODS MCLA-128 concentration data following intravenous administration were collected in a phase I/II clinical trial. Pharmacokinetic data were analyzed using non-linear mixed-effects modeling. Different compartmental models were evaluated. Various body size parameters including body weight, body surface area, and fat-free mass were evaluated as covariates in addition to age, sex, HER2 status, and tumor burden. RESULTS In total, 1115 serum concentration measurements were available from 116 patients. The pharmacokinetics of MCLA-128 was best described by a two-compartment model with linear and non-linear (Michaelis-Menten) clearance. Fat-free mass significantly affected the linear clearance and volume of distribution of the central compartment of MCLA-128, explaining 8.4% and 5.6% of inter-individual variability, respectively. Tumor burden significantly affected the non-linear clearance capacity. Simulations demonstrated that dosing based on body size parameters resulted in similar area under the plasma concentration-time curve for a dosing interval (AUC0-τ), maximum and trough concentrations of MCLA-128, compared to flat dosing. CONCLUSIONS This analysis demonstrated that the pharmacokinetics of MCLA-128 exhibits similar disposition characteristics to other therapeutic monoclonal antibodies and that a flat dose of MCLA-128 in patients with various solid tumors is appropriate.
Collapse
|
13
|
Understanding Inter-Individual Variability in Monoclonal Antibody Disposition. Antibodies (Basel) 2019; 8:antib8040056. [PMID: 31817205 PMCID: PMC6963779 DOI: 10.3390/antib8040056] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 12/29/2022] Open
Abstract
Monoclonal antibodies (mAbs) are currently the largest and most dominant class of therapeutic proteins. Inter-individual variability has been observed for several mAbs; however, an understanding of the underlying mechanisms and factors contributing to inter-subject differences in mAb disposition is still lacking. In this review, we analyze the mechanisms of antibody disposition and the putative mechanistic determinants of inter-individual variability. Results from in vitro, preclinical, and clinical studies were reviewed evaluate the role of the neonatal Fc receptor and Fc gamma receptors (expression and polymorphism), target properties (expression, shedding, turnover, internalization, heterogeneity, polymorphism), and the influence of anti-drug antibodies. Particular attention is given to the influence of co-administered drugs and disease, and to the physiological relevance of covariates identified by population pharmacokinetic modeling, as determinants of variability in mAb pharmacokinetics.
Collapse
|
14
|
Krasniqi E, Barchiesi G, Pizzuti L, Mazzotta M, Venuti A, Maugeri-Saccà M, Sanguineti G, Massimiani G, Sergi D, Carpano S, Marchetti P, Tomao S, Gamucci T, De Maria R, Tomao F, Natoli C, Tinari N, Ciliberto G, Barba M, Vici P. Immunotherapy in HER2-positive breast cancer: state of the art and future perspectives. J Hematol Oncol 2019; 12:111. [PMID: 31665051 PMCID: PMC6820969 DOI: 10.1186/s13045-019-0798-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/25/2019] [Indexed: 02/08/2023] Open
Abstract
Breast cancer (BC) is a complex disease with primary or acquired incurability characteristics in a significant part of patients. Immunotherapeutical agents represent an emerging option for breast cancer treatment, including the human epidermal growth factor 2 positive (HER2+) subtype. The immune system holds the ability to spontaneously implement a defensive response against HER2+ BC cells through complex mechanisms which can be exploited to modulate this response for obtaining a clinical benefit. Initial immune system modulating strategies consisted mostly in vaccine therapies, which are still being investigated and improved. However, the entrance of trastuzumab into the scenery of HER2+ BC treatment was the real game changing event, which embodied a dominant immune-mediated mechanism. More recently, the advent of the immune checkpoint inhibitors has caused a new paradigm shift for immuno-oncology, with promising initial results also for HER2+ BC. Breast cancer has been traditionally considered poorly immunogenic, being characterized by relatively low tumor mutation burden (TMB). Nevertheless, recent evidence has revealed high tumor infiltrating lymphocytes (TILs) and programmed cell death-ligand 1 (PD-L1) expression in a considerable proportion of HER2+ BC patients. This may translate into a higher potential to elicit anti-cancer response and, therefore, wider possibilities for the use and implementation of immunotherapy in this subset of BC patients. We are herein presenting and critically discussing the most representative evidence concerning immunotherapy in HER2+ BC cancer, both singularly and in combination with therapeutic agents acting throughout HER2-block, immune checkpoint inhibition and anti-cancer vaccines. The reader will be also provided with hints concerning potential future projection of the most promising immutherapeutic agents and approaches for the disease of interest.
Collapse
Affiliation(s)
- E Krasniqi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - G Barchiesi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - L Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - M Mazzotta
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy
| | - A Venuti
- HPV-UNIT, UOSD Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostic and Technological Innovation (RIDAIT), Translational Research Functional Departmental Area, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - M Maugeri-Saccà
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - G Sanguineti
- Department of Radiation Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - G Massimiani
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - D Sergi
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - S Carpano
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| | - P Marchetti
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, Azienda Ospedaliera Sant'Andrea, Rome, Italy.,Medical Oncology Unit B, Policlinico Umberto I, Rome, Italy
| | - S Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Policlinico Umberto I, 'Sapienza' University of Rome, Rome, Italy
| | - T Gamucci
- Medical Oncology, Sandro Pertini Hospital, Rome, Italy
| | - R De Maria
- Institute of General Pathology, Catholic University of the Sacred Heart, Rome, Italy.,Department of Medical Oncology, Policlinico Universitario "A. Gemelli", Rome, Italy
| | - F Tomao
- Department of Gynecology-Obstetrics and Urology, "Sapienza" University of Rome, Rome, Italy
| | - C Natoli
- Department of Medical, Oral and Biotechnological Sciences and Center of Aging Science & Translational Medicine (CeSI-MeT), G. d'Annunzio University, Chieti, Italy
| | - N Tinari
- Department of Medical, Oral and Biotechnological Sciences and Center of Aging Science & Translational Medicine (CeSI-MeT), G. d'Annunzio University, Chieti, Italy
| | - G Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - M Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy.
| | - P Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53-00144, Rome, Italy
| |
Collapse
|
15
|
Bartelink IH, Jones EF, Shahidi‐Latham SK, Lee PRE, Zheng Y, Vicini P, van ‘t Veer L, Wolf D, Iagaru A, Kroetz DL, Prideaux B, Cilliers C, Thurber GM, Wimana Z, Gebhart G. Tumor Drug Penetration Measurements Could Be the Neglected Piece of the Personalized Cancer Treatment Puzzle. Clin Pharmacol Ther 2019; 106:148-163. [PMID: 30107040 PMCID: PMC6617978 DOI: 10.1002/cpt.1211] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/30/2018] [Indexed: 12/30/2022]
Abstract
Precision medicine aims to use patient genomic, epigenomic, specific drug dose, and other data to define disease patterns that may potentially lead to an improved treatment outcome. Personalized dosing regimens based on tumor drug penetration can play a critical role in this approach. State-of-the-art techniques to measure tumor drug penetration focus on systemic exposure, tissue penetration, cellular or molecular engagement, and expression of pharmacological activity. Using in silico methods, this information can be integrated to bridge the gap between the therapeutic regimen and the pharmacological link with clinical outcome. These methodologies are described, and challenges ahead are discussed. Supported by many examples, this review shows how the combination of these techniques provides enhanced patient-specific information on drug accessibility at the tumor tissue level, target binding, and downstream pharmacology. Our vision of how to apply tumor drug penetration measurements offers a roadmap for the clinical implementation of precision dosing.
Collapse
Affiliation(s)
- Imke H. Bartelink
- Department of MedicineUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneSouth San FranciscoCaliforniaUSA
- Department of Clinical Pharmacology and PharmacyAmsterdam UMCVrije Universiteit AmsterdamThe Netherlands
| | - Ella F. Jones
- Department of Radiology and Biomedical ImagingUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Pei Rong Evelyn Lee
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Yanan Zheng
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneSouth San FranciscoCaliforniaUSA
| | - Paolo Vicini
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD)MedImmuneCambridgeUK
| | - Laura van ‘t Veer
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Denise Wolf
- Department of Laboratory Medicine of the UCSF Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging at Stanford Health CareStanfordCaliforniaUSA
| | - Deanna L. Kroetz
- Department of Bioengineering and Therapeutic Sciences (BTS)School of PharmacyUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Brendan Prideaux
- Rutgers New Jersey Medical SchoolPublic Health Research InstituteRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Cornelius Cilliers
- Departments of Chemical Engineering and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Greg M. Thurber
- Departments of Chemical Engineering and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Zena Wimana
- Institut Jules BordetUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| | - Geraldine Gebhart
- Institut Jules BordetUniversité Libre de Bruxelles (ULB)BrusselsBelgium
| |
Collapse
|
16
|
Nguyen TT, Angeli E, Darrouzain F, Nguyen QT, Desvignes C, Rigal M, Nevine O, Nicolas P, Le QV, Winterman S, Pailler MC, Zelek L, Paintaud G, Janin A, Bousquet G. A successful compartmental approach for the treatment of breast cancer brain metastases. Cancer Chemother Pharmacol 2019; 83:573-580. [PMID: 30610367 DOI: 10.1007/s00280-018-3752-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/04/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Brain metastases are challenging daily practice in oncology and remain a compartmental problem since most anti-cancer drugs do not cross the blood-brain barrier at relevant pharmacological concentrations. METHODS In a young woman with HER2-overexpressing breast cancer resistant to standard treatments, at the time of brain metastases progression, a ventricular reservoir was implanted for intrathecal drug injections and detailed pharmacokinetic studies. RESULTS A first association of intrathecal trastuzumab with intravenous cisplatin was offered to the patient. For trastuzumab, the mean cerebrospinal fluid trough concentration of 53.4 mg/L reached relevant levels, enabling the stabilization of the metastases. Adding intravenous cisplatin was not beneficial, since the cerebrospinal fluid exposure was almost undetectable under 0.08 mg/L. We then offered the patient an intrathecal combination of trastuzumab and methotrexate, because of their in vitro synergic cytotoxicity. The cerebrospinal fluid peak of methotrexate was 1037 µmol/L at 2 h, and the concentrations remained above the theoretical therapeutic concentration. After 2 months of this drug combination, we obtained an excellent response on the brain metastases. CONCLUSION Our preliminary study supports the interest of a compartmental approach through a direct administration of drugs into the cerebrospinal fluid for the treatment of breast cancer brain metastases.
Collapse
Affiliation(s)
- Thuy T Nguyen
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France.,AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France.,Medical Oncology Department A, National Cancer Hospital, Ha Noi, Viet Nam
| | - Eurydice Angeli
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France
| | - François Darrouzain
- Université François-Rabelais de Tours, CNRS, GICC UMR 7292, 37000, Tours, France.,CHRU de Tours, Pharmacologie-Toxicologie, 37000, Tours, France
| | - Quang T Nguyen
- Medical Oncology Department A, National Cancer Hospital, Ha Noi, Viet Nam
| | | | - Marthe Rigal
- AP-HP Hôpital Avicenne, Pharmacie, 93000, Bobigny, France
| | - Osman Nevine
- AP-HP Hôpital Avicenne, Pharmacie, 93000, Bobigny, France
| | - Patrick Nicolas
- AP-HP Hôpital Avicenne, Service de Biochimie, 93000, Bobigny, France
| | - Quang V Le
- Surgery Department A, National Cancer Hospital, Ha Noi, Viet Nam
| | - Sabine Winterman
- AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France
| | | | - Laurent Zelek
- AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France.,Université Paris 13, 93430, Villetaneuse, France
| | - Gilles Paintaud
- Université François-Rabelais de Tours, CNRS, GICC UMR 7292, 37000, Tours, France.,CHRU de Tours, Pharmacologie-Toxicologie, 37000, Tours, France
| | - Anne Janin
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France. .,AP-HP Hôpital Saint-Louis, Laboratoire de Pathologie, 75010, Paris, France. .,Université Paris Diderot, Sorbonne Paris Cité, UMR-S 1165, 75010, Paris, France.
| | - Guilhem Bousquet
- U1165, Université Paris 7, INSERM, Hôpital Saint-Louis, 1 Avenue Claude Vellefaux, 75010, Paris, France. .,AP-HP Hôpital Avicenne, Service d'Oncologie Médicale, 93000, Bobigny, France. .,Université Paris 13, 93430, Villetaneuse, France. .,Université Paris Diderot, Sorbonne Paris Cité, UMR-S 1165, 75010, Paris, France.
| |
Collapse
|
17
|
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
|
18
|
A novel way to manage trastuzumab cardiotoxicity. Cancer Chemother Pharmacol 2018; 81:791-796. [PMID: 29497813 DOI: 10.1007/s00280-018-3555-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 02/27/2018] [Indexed: 10/17/2022]
Abstract
PURPOSE Trastuzumab is the most widely prescribed anti-HER2 humanized monoclonal antibody. Cardiac toxicity is the only limiting toxicity of trastuzumab and it is of particular concern in patients with complete response, since the drug needs to be stopped, with a risk of disease relapse. To date, no pharmacological data on trastuzumab cardiotoxicity in patients have been made available. Here, we provide proof of concept, demonstrating that it was possible to prevent trastuzumab-induced cardiotoxicity by modifying the drug administration schedule. METHODS In this paper, we report the case of a patient with metastatic breast cancer responding to trastuzumab, who developed severe cardiac toxicity twice using a 3-weekly regimen. Considering preclinical pharmacological data on trastuzumab cardiotoxicity, we hypothesized that a weekly schedule of trastuzumab with lower peaks of serum concentration could be safe while remaining efficient. With the patient's consent, we started a weekly combination of carboplatin (AUC2) and trastuzumab (2 mg/kg) with close monitoring of trastuzumab concentrations. RESULTS We successfully controlled the disease for an additional 6 months with relevant trough concentrations of trastuzumab of around 50 mg/L. Another important aspect is that, with this weekly schedule, we observed no cardiac toxicity, and the left ventricular ejection fraction remained stabilized, at over 50%. CONCLUSIONS Trastuzumab is the most widely prescribed anti-HER2 monoclonal antibody for the treatment of HER2 metastatic breast cancer, and it is the only drug that has been approved for the treatment of localized HER2 breast cancer, 1-year treatment being required after surgery. In case of cardiac toxicity, particularly in women over 60 years of age, a weekly regimen with lower peaks of concentration could be an alternative to the standard 3-weekly regimen.
Collapse
|
19
|
Bevacizumab Pharmacokinetics Influence Overall and Progression-Free Survival in Metastatic Colorectal Cancer Patients. Clin Pharmacokinet 2017; 55:1381-1394. [PMID: 27312193 DOI: 10.1007/s40262-016-0406-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Clinical response to bevacizumab varies between patients treated for metastatic colorectal cancer (mCRC). The aim of this study was to quantify individual factors affecting bevacizumab pharmacokinetic variability and assess the relationship between bevacizumab concentrations and clinical outcomes. METHODS Bevacizumab pharmacokinetics were assessed in 130 mCRC patients using a two-compartment pharmacokinetic population model. Overall and progression-free survival (PFS) were analyzed using Cox models. RESULTS The bevacizumab volume of distribution increased with height (p = 10-10) and was higher in patients with a 3/3 variable number tandem repeat of the FCGRT (Fc fragment of IgG receptor and transporter) gene (p = 0.039). The elimination rate constant increased with baseline carcinoembryonic antigen (CEA) and vascular endothelial growth factor (VEGF) concentrations, and was higher in patients with extra-hepatic metastases (p = 0.00029, 0.011, and 0.014). A bevacizumab trough concentration ≤15.5 mg/L was associated with both shorter overall survival and PFS (hazard ratio [95 % CI] 1.90 [1.20-2.99] and 1.76 [1.20-2.58], respectively). CONCLUSION High tumour burden is associated with low bevacizumab concentrations, and high bevacizumab concentration are associated with both decreased overall and progression-free survivals.
Collapse
|
20
|
Singh AP, Shah DK. Application of a PK-PD Modeling and Simulation-Based Strategy for Clinical Translation of Antibody-Drug Conjugates: a Case Study with Trastuzumab Emtansine (T-DM1). AAPS JOURNAL 2017; 19:1054-1070. [PMID: 28374319 DOI: 10.1208/s12248-017-0071-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/28/2017] [Indexed: 02/06/2023]
Abstract
Successful clinical translation of antibody-drug conjugates (ADCs) can be challenging due to complex pharmacokinetics and differences between preclinical and clinical tumors. To facilitate this translation, we have developed a general pharmacokinetic-pharmacodynamic (PK-PD) modeling and simulation (M&S)-based strategy for ADCs. Here we present the validation of this strategy using T-DM1 as a case study. A previously developed preclinical tumor disposition model for T-DM1 (Singh and Shah, AAPSJ. 2015; 18(4):861-875) was used to develop a PK-PD model that can characterize in vivo efficacy of T-DM1 in preclinical tumor models. The preclinical data was used to estimate the efficacy parameters for T-DM1. Human PK of T-DM1 was a priori predicted using allometric scaling of monkey PK parameters. The predicted human PK, preclinically estimated efficacy parameters, and clinically observed volume and growth parameters for breast cancer were combined to develop a translated clinical PK-PD model for T-DM1. Clinical trial simulations were performed using the translated PK-PD model to predict progression-free survival (PFS) and objective response rates (ORRs) for T-DM1. The model simulated PFS rates for HER2 1+ and 3+ populations were comparable to the rates observed in three different clinical trials. The model predicted only a modest improvement in ORR with an increase in clinically approved dose of T-DM1. However, the model suggested that a fractionated dosing regimen (e.g., front loading) may provide an improvement in the efficacy. In general, the PK-PD M&S-based strategy presented here is capable of a priori predicting the clinical efficacy of ADCs, and this strategy has been now retrospectively validated for all clinically approved ADCs.
Collapse
Affiliation(s)
- Aman P Singh
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, New York, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York at Buffalo, 455 Kapoor Hall, Buffalo, New York, 14214-8033, USA.
| |
Collapse
|
21
|
Abstract
Monoclonal antibodies (MAbs) have become a substantial part of many pharmaceutical company portfolios. However, the development process of MAbs for clinical use is quite different than for small-molecule drugs. MAb development programs require careful interdisciplinary evaluations to ensure the pharmacology of both the MAb and the target antigen are well-understood. Selection of appropriate preclinical species must be carefully considered and the potential development of anti-drug antibodies (ADA) during these early studies can limit the value and complicate the performance and possible duration of preclinical studies. In human studies, many of the typical pharmacology studies such as renal or hepatic impairment evaluations may not be needed but the pharmacokinetics and pharmacodynamics of these agents is complex, often necessitating more comprehensive evaluation of clinical data and more complex bioanalytical assays than might be used for small molecules. This paper outlines concerns and strategies for development of MAbs from the early in vitro assessments needed through preclinical and clinical development. This review focuses on how to develop, submit, and comply with regulatory requirements for MAb therapeutics.
Collapse
|
22
|
Tout M, Passot C, Cartron G, Paintaud G, Ternant D. Gota et al. on their article "the pharmacokinetics of Reditux™, a biosimilar of rituximab". Cancer Chemother Pharmacol 2016; 78:1317-1318. [PMID: 27752763 DOI: 10.1007/s00280-016-3176-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/12/2016] [Indexed: 11/24/2022]
Affiliation(s)
- Mira Tout
- CNRS, GICC UMR 7292, Université François-Rabelais de Tours, Tours, France
| | - Christophe Passot
- CNRS, GICC UMR 7292, Université François-Rabelais de Tours, Tours, France.,Laboratoire de Pharmacologie-Toxicologie, CHU de Tours, 2 Boulevard Tonnellé, 37044, Tours Cedex, France
| | - Guillaume Cartron
- CNRS, UMR 5235, Université de Montpellier, Montpellier, France.,Department of Hematology, CHU Montpellier, Montpellier, France
| | - Gilles Paintaud
- CNRS, GICC UMR 7292, Université François-Rabelais de Tours, Tours, France.,Laboratoire de Pharmacologie-Toxicologie, CHU de Tours, 2 Boulevard Tonnellé, 37044, Tours Cedex, France
| | - David Ternant
- CNRS, GICC UMR 7292, Université François-Rabelais de Tours, Tours, France. .,Laboratoire de Pharmacologie-Toxicologie, CHU de Tours, 2 Boulevard Tonnellé, 37044, Tours Cedex, France.
| |
Collapse
|
23
|
Pharmacokinetics interactions of monoclonal antibodies. Pharmacol Res 2016; 111:592-599. [DOI: 10.1016/j.phrs.2016.07.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 12/27/2022]
|
24
|
Bernadou G, Campone M, Merlin JL, Gouilleux-Gruart V, Bachelot T, Lokiec F, Rezai K, Arnedos M, Diéras V, Jimenez M, Paintaud G, Ternant D. Influence of tumour burden on trastuzumab pharmacokinetics in HER2 positive non-metastatic breast cancer. Br J Clin Pharmacol 2016; 81:941-8. [PMID: 26714164 DOI: 10.1111/bcp.12875] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/01/2015] [Accepted: 12/24/2015] [Indexed: 01/15/2023] Open
Abstract
AIMS Trastuzumab, an antibody binding to epidermal growth factor receptor-2 (HER2), has been approved to treat HER2-positive breast cancer in different settings. This study aimed at evaluating the influence of tumour size on trastuzumab pharmacokinetics (PK) in non-metastatic breast cancer patients treated with short term pre-operative trastuzumab. METHODS Trastuzumab PK data were obtained from a multicentre, randomized and comparative study. This antibody was administered pre-operatively to patients with localized HER2-positive breast cancer as a single 4 mg kg(-1) loading dose followed by 5 weekly 2 mg kg(-1) doses. Trastuzumab concentrations were measured repeatedly using an ELISA technique. Tumour size was evaluated at baseline using breast echography. Trastuzumab pharmacokinetics were studied using a population approach and a two compartment model. The influence of tumour burden on trastuzumab pharmacokinetics was quantified as a covariate. RESULTS A total of 784 trastuzumab concentrations were available from the 79 eligible patients. Estimated parameters (interindiviual standard deviation) were central volume of distribution =2.1 l (23%), peripheral volume of distribution =1.3 l (38%), intercompartment clearance =0.36 l day(-1) , with an elimination half-life of 11.8 days. Typical clearance was 0.22 l day(-1) (19%) and its value was increased with tumour size. In patients with the highest tumour size, trastuzumab clearance was 50% [18%-92%] higher than in patients with the lowest tumour size. CONCLUSIONS In non-metastatic breast cancer patients, trastuzumab clearance increases with tumour size. The elimination half-life of trastuzumab was shorter in the present population of patients than in metastatic breast cancer patients previously studied.
Collapse
Affiliation(s)
- Guillemette Bernadou
- CNRS, UMR 7292, « Genetics, Immunotherapy, Chemistry and Cancer », Université François Rabelais de Tours, Tours.,CHRU de Tours, Clinique d'Oncologie et de Radiothérapie, Tours
| | - Mario Campone
- Department of Medical Oncology/Cancer Research Center UMR-INSERM, U892/CNRS 6299/Bioinformatics Unit, Institut de Cancérologie de l'Ouest, Nantes
| | - Jean-Louis Merlin
- Faculté de Pharmacie, Université de Lorraine, Nancy.,CNRS UMR7039 CRAN, Vandoeuvre les Nancy.,Service de Biopathologie, Institut de Cancérologie de Lorraine, Vandoeuvre les Nancy
| | - Valérie Gouilleux-Gruart
- CNRS, UMR 7292, « Genetics, Immunotherapy, Chemistry and Cancer », Université François Rabelais de Tours, Tours.,CHRU de Tours, Service d'Immunologie, Tours
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, INSERM U1052, Lyon
| | - François Lokiec
- Department of Radiopharmacology, Institut Curie-René Huguenin Hospital, Saint-Cloud
| | - Keyvan Rezai
- Department of Radiopharmacology, Institut Curie-René Huguenin Hospital, Saint-Cloud
| | - Monica Arnedos
- Department of Medical Oncology, Gustave Roussy, Villejuif
| | | | | | - Gilles Paintaud
- CNRS, UMR 7292, « Genetics, Immunotherapy, Chemistry and Cancer », Université François Rabelais de Tours, Tours.,CHRU de Tours, Laboratoire de Pharmacologie-Toxicologie, Tours, France
| | - David Ternant
- CNRS, UMR 7292, « Genetics, Immunotherapy, Chemistry and Cancer », Université François Rabelais de Tours, Tours.,CHRU de Tours, Laboratoire de Pharmacologie-Toxicologie, Tours, France
| |
Collapse
|