1
|
Chan P, Liu SN, Gosselin N, Sauve Z, Marchand M, Lin A, Herraez‐Baranda L, Zanghi J, Shearer‐Kang E, Liu X, Wu B, Chanu P. Population Pharmacokinetics and Exposure-Response of Subcutaneous Atezolizumab in Patients With Non-Small Cell Lung Cancer. CPT Pharmacometrics Syst Pharmacol 2025; 14:726-737. [PMID: 39906948 PMCID: PMC12001254 DOI: 10.1002/psp4.13310] [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: 11/18/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
IMscin001 is a two-part dose-finding (Phase Ib) and -confirmation (Phase III) study to evaluate atezolizumab pharmacokinetics of subcutaneous (SC) compared with intravenous (IV) administration in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC). The objectives of the current analyses were to characterize the population pharmacokinetics (popPK) of atezolizumab and to determine the relationship between atezolizumab exposure and safety and efficacy endpoints in IMscin001. A previously validated IV popPK model was extended to add SC absorption parameters using SC and IV data from Phase Ib and Phase III of IMscin001 (N = 435), and covariate effects were investigated on the SC absorption parameters. The exposure-response (ER) investigation was performed using SC data following 1875 mg every three weeks (q3w) administration in the Phase III portion of IMscin001 (N = 246). The clinical endpoints were objective response rate, progression-free survival, or overall survival for efficacy, serious adverse events, special interest adverse events, Grades 3-5 adverse events, infusion-related reaction, or injection site reactions for safety. Atezolizumab SC absorption was characterized by a first-order absorption with a bioavailability of 71.8% and an absorption rate constant of 0.304 day-1. The extended popPK model was adequate to predict atezolizumab PK after IV and SC administrations and to predict individual exposure metrics. For all efficacy and safety endpoints, atezolizumab exposure was not statistically significant (p-value > 0.05) in the ER models. The non-inferior popPK exposure and flat ER results supported atezolizumab SC dose at 1875 mg q3w.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Lung Neoplasms/drug therapy
- Male
- Female
- Injections, Subcutaneous
- Middle Aged
- Aged
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Dose-Response Relationship, Drug
- Models, Biological
- Adult
- Administration, Intravenous
Collapse
|
2
|
Agema BC, Koch BCP, Mathijssen RHJ, Koolen SLW. From Prospective Evaluation to Practice: Model-Informed Dose Optimization in Oncology. Drugs 2025; 85:487-503. [PMID: 39939511 PMCID: PMC11946950 DOI: 10.1007/s40265-025-02152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2025] [Indexed: 02/14/2025]
Abstract
One dose does not fit all, especially in oncolytic drugs, where side effects and therapy failures highlight the need for personalized dosing approaches. In recent years, the quest to apply model-informed precision dosing to oncology drugs has gained significant momentum, reflecting its potential to revolutionize patient care by tailoring treatments to individual pharmacokinetic profiles. Despite this progress, model-informed precision dosing has not (yet) become widely integrated into routine clinical care. We aimed to explain model-informed precision dosing from a clinical viewpoint while addressing all prospective model-informed precision dosing implementation and validation studies in the field of oncology. We identified 16 different drugs for which prospective model-informed precision dosing validation/implementation has been performed. Although these studies are mostly focused on attaining adequate drug exposures and reducing inter-individual variability, improved clinical outcomes after performing model-informed precision dosing were shown for busulfan, and high-dose methotrexate. Toxicities were significantly reduced for busulfan and cyclophosphamide treatment. In contrast, for carboplatin, for which model-informed precision dosing has been used in the Calvert formula, no prospective validation on outcomes was deemed necessary as the therapeutic window had been extensively validated. Model-informed precision dosing has shown to be of added value in oncology and is expected to significantly change dosing regimens in the future.
Collapse
Affiliation(s)
- Bram C Agema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
- Rotterdam Clinical Pharmacometrics Group, Rotterdam, The Netherlands.
| | - Birgit C P Koch
- Rotterdam Clinical Pharmacometrics Group, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Hartung N, Khatova A. Information-theoretic evaluation of covariate distributions models. J Pharmacokinet Pharmacodyn 2025; 52:21. [PMID: 40148687 PMCID: PMC11950120 DOI: 10.1007/s10928-025-09968-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
Statistical modelling of covariate distributions allows to generate virtual populations or to impute missing values in a covariate dataset. Covariate distributions typically have non-Gaussian margins and show nonlinear correlation structures, which simple descriptions like multivariate Gaussian distributions fail to represent. Prominent non-Gaussian frameworks for covariate distribution modelling are copula-based models and models based on multiple imputation by chained equations (MICE). While both frameworks have already found applications in the life sciences, a systematic investigation of their goodness-of-fit to the theoretical underlying distribution, indicating strengths and weaknesses under different conditions, is still lacking. To bridge this gap, we thoroughly evaluated covariate distribution models in terms of Kullback-Leibler (KL) divergence, a scale-invariant information-theoretic goodness-of-fit criterion for distributions. Methodologically, we proposed a new approach to construct confidence intervals for KL divergence by combining nearest neighbour-based KL divergence estimators with subsampling-based uncertainty quantification. In relevant data sets of different sizes and dimensionalities with both continuous and discrete covariates, non-Gaussian models showed consistent improvements in KL divergence, compared to simpler Gaussian or scale transform approximations. KL divergence estimates were also robust to the inclusion of latent variables and large fractions of missing values. While good generalization behaviour to new data could be seen in copula-based models, MICE shows a trend for overfitting and its performance should always be evaluated on separate test data. Parametric copula models and MICE were found to scale much better with the dimension of the dataset than nonparametric copula models. These findings corroborate the potential of non-Gaussian models for modelling realistic life science covariate distributions.
Collapse
Affiliation(s)
- Niklas Hartung
- Institute of Mathematics, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam, Germany.
| | - Aleksandra Khatova
- Institute of Mathematics, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476, Potsdam, Germany
- Faculty of Health Sciences, BTU Cottbus-Senftenberg, Lipezker Straße 47, 03048, Cottbus, Germany
| |
Collapse
|
4
|
Canzian J, Conforti F, Jacobs F, Benvenuti C, Gaudio M, Gerosa R, De Sanctis R, Zambelli A. Sex-Related Differences in Immunotherapy Toxicities: Insights into Dimorphic Responses. Cancers (Basel) 2025; 17:1054. [PMID: 40227458 PMCID: PMC11987764 DOI: 10.3390/cancers17071054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Significant sex-based differences exist in the immune system and antitumor immune responses, potentially leading to variations in both the efficacy and toxicity of anticancer immunotherapies. Women generally mount stronger innate and adaptive immune responses than men, which can result in more severe immune-related adverse events (irAEs) during treatments with immune checkpoint inhibitors (ICIs). However, the importance of sex dimorphism in the safety of cancer immunotherapy remains underexplored in clinical oncology, despite its profound implications for treatment outcomes. Our review highlights the critical influence of biological sex on pharmacokinetics, pharmacodynamics, and immune responses, shaping ICI efficacy and the prevalence, type, and severity of irAEs. Integrating sex as a critical variable in cancer treatment and clinical trial design is essential for personalizing therapeutic strategies, bridging existing knowledge gaps, and enhancing survival rates and quality of life for patients across genders.
Collapse
Affiliation(s)
- Jacopo Canzian
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Fabio Conforti
- Division of Medical Oncology, Humanitas Gavazzeni, 24125 Bergamo, Italy;
| | - Flavia Jacobs
- Division of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Chiara Benvenuti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Mariangela Gaudio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Riccardo Gerosa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Rita De Sanctis
- Oncology Unit, ASST Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy;
| | - Alberto Zambelli
- Oncology Unit, ASST Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy;
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
5
|
Vitale E, Maistrello L, Rizzo A, Brunetti O, Massafra R, Mollica V, Massari F, Santoni M. Nutritional conditions and PFS and OS in cancer immunotherapy: the MOUSEION-010 meta-analysis. Immunotherapy 2025; 17:269-281. [PMID: 40134096 PMCID: PMC12013447 DOI: 10.1080/1750743x.2025.2483656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The MOUSEION-010 Meta-Analysis assessed the association between nutritional status and clinical outcomes such as Progression Free Survival (PFS) and Overall Survival (OS) among cancer patients treated with immune checkpoint inhibitors (ICIs). METHODS Nutritional status was assessed based on the Prognostic Nutrition Index (PNI), Geriatric Nutritional Risk Index (GNRI) and Controlling Nutritional Status (CONUT) indexes. Databases consulted were: Embase, PubMed, Scopus and Web of Science. RESULTS PNI and GNRI indexes did not show a significant association with both PFS and OS, while CONUT index displayed a significant difference in PFS between the two groups, in favor of the control group (Z = 4.04; p < 0.01) also without any publication bias (β= -1.27; 95% CI = [-2.13; -0.42]; p = 0.10]). The same trend was recorded in OS, too (Z = 4.24; p < 0.01). However, publication bias was present (β = 1.89; 95% CI = [1.26; 2.54]; p = 0.028]) and the numerosity of the studies did not reveal the sufficient statistical power to obtain reliable results. CONCLUSION Malnutrition could negatively impact cancer patients, especially in advanced phases. Our findings could be associated with the reduction of physical ability and daily activity performance, lower compliance with treatment protocols, and shorter survival outcomes.
Collapse
Affiliation(s)
- Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | | | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | |
Collapse
|
6
|
Maritaz C, Combarel D, Dalban C, Blondel L, Broutin S, Marabelle A, Albiges L, Paci A. Nivolumab plasma concentration and clearance associated with overall survival in patients with renal cell carcinoma. J Immunother Cancer 2025; 13:e010059. [PMID: 39762076 PMCID: PMC11749330 DOI: 10.1136/jitc-2024-010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/23/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Nivolumab is an immune checkpoint inhibitor (ICI) that selectively inhibits programmed cell death protein 1 activation, restoring antitumor immunity. ICIs are indicated for various types of advanced solid tumors; however, not all patients benefit from them, and tools that could be used in the clinic to predict response to treatment represent an unmet need. Here we describe the development of a new population pharmacokinetic (PPK) model in patients treated with nivolumab in clinical trials. Applying the model to a patient population with renal cell carcinoma identified nivolumab clearance and plasma concentration as predictors of overall survival (OS). METHODS A custom liquid chromatography with tandem mass spectrometry method for quantifying nivolumab plasma concentration was developed and validated following the European Medicines Agency guidelines for bioanalytical method validation. The PPK model was developed using data from patients treated in the NIVIPIT (n=38) and NIVOREN (n=137) trials of nivolumab in metastatic melanoma and renal cell carcinoma, respectively. The PPK model was used to determine pharmacokinetic (PK) parameters such as baseline clearance and simulate individual clearance changes over time. The relationship between PK characteristics (including clearance at Cycle 1 (CLC1), plasma concentration at Cycle 3 and clinical outcomes was assessed in 137 patients treated in NIVOREN. Kaplan-Meier methodology was used in time-to-event analyses. RESULTS In 137 patients, the median nivolumab CLC1 was 6 mL/hour and the median plasma concentration at Cycle 3 was 48 µg/mL. Median follow-up was 21.0 months (95% CI 20.2 to 22.5 months) with a survival rate at 6 months of 91.2% and 77.9% at 12 months. In univariate analysis, OS was significantly higher in patients with CLC1<6 mL/hour versus ≥6 mL/hour (HR 2.2 (95% CI 1.2 to 4.1), p=0.0146). Shorter OS was observed in patients with plasma concentration at Cycle 3 below the median (48 µg/mL) versus those above the median (HR 0.4 (95% CI 0.2 to 0.8), p=0.0069). Multivariate analysis showed a trend towards lower clearance, but this did not reach statistical significance (p=0.0694). CONCLUSIONS Results of the study may potentially be used to predict outcomes of nivolumab therapy in patients with renal cell carcinoma. Additional applications may include guiding dose adjustments of nivolumab in those who are less likely to respond to the initial dose.
Collapse
Affiliation(s)
- Christophe Maritaz
- Paris-Saclay University, Gif-sur-Yvette, Île-de-France, France
- Gustave Roussy, Villejuif, Île-de-France, France
- Pharmacology Department, Gustave Roussy, Villejuif, Île-de-France, France
| | - David Combarel
- Paris-Saclay University, Gif-sur-Yvette, Île-de-France, France
- Gustave Roussy, Villejuif, Île-de-France, France
- Pharmacology Department, Gustave Roussy, Villejuif, Île-de-France, France
| | - Cécile Dalban
- Biostatistics Department, Centre Leon Berard, Lyon, Auvergne-Rhône-Alpes, France
| | | | | | | | | | - Angelo Paci
- Paris-Saclay University, Gif-sur-Yvette, Île-de-France, France
- Gustave Roussy, Villejuif, Île-de-France, France
- Pharmacology Department, Gustave Roussy, Villejuif, Île-de-France, France
| |
Collapse
|
7
|
Majid O, Cao Y, Willis BA, Hayato S, Takenaka O, Lalovic B, Sreerama Reddy SH, Penner N, Reyderman L, Yasuda S, Hussein Z. Population pharmacokinetics and exposure-response analyses of safety (ARIA-E and isolated ARIA-H) of lecanemab in subjects with early Alzheimer's disease. CPT Pharmacometrics Syst Pharmacol 2024; 13:2111-2123. [PMID: 39207112 PMCID: PMC11646937 DOI: 10.1002/psp4.13224] [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: 02/28/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Lecanemab (Leqembi®) was recently approved by health authorities in the United States, Japan, and China to treat early Alzheimer's disease (AD), including patients with mild cognitive impairment (MCI) or mild dementia due to Alzheimer's disease upon confirmation of amyloid beta pathology. Extensively and sparsely sampled PK profiles from 1619 AD subjects and 21,929 serum lecanemab observations from two phase I, one phase II, and one phase III studies were well characterized using a two-compartment model with first-order elimination. The final PK model quantified covariate effects of body weight and sex on clearance and central volume of distribution, ADA-positive status, and albumin on clearance, and of Japanese ethnicity on central and peripheral volumes of distribution. Exposure to lecanemab was comparable between two lecanemab-manufacturing processes. However, none of the identified covariates in the model had a clinically relevant impact on model-predicted lecanemab Cmax or AUC at steady state following 10 mg/kg bi-weekly. Importantly, age, a well-recognized risk factor for AD, was not found to significantly affect lecanemab PK. The incidence of ARIA-E as a function of lecanemab exposure was modeled using a logit function with data pooled from 2641 subjects from the phase II and phase III studies, in which a total of 177 incidences of ARIA-E were observed. The probability of ARIA-E was significantly correlated with model-predicted Cmax and predicted to be higher in subjects homozygous for APOE4. The incidence of isolated ARIA-H was not associated with lecanemab exposure and was similar between placebo and lecanemab-treated subjects.
Collapse
|
8
|
Vogg B, Poetzl J, Schwebig A, Sekhar S, Kivitz A, Krivtsova N, Renner O, Body JJ, Eastell R. The Totality of Evidence for SDZ-deno: A Biosimilar to Reference Denosumab. Clin Ther 2024; 46:916-926. [PMID: 39294041 DOI: 10.1016/j.clinthera.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/12/2024] [Indexed: 09/20/2024]
Abstract
PURPOSE Sandoz biosimilar denosumab (GP2411 [SDZ-deno]; Jubbonti/Wyost) is approved by the US FDA, EMA and Health Canada for all indications of reference denosumab (REF-deno; Prolia/Xgeva), a fully human IgG2κ monoclonal antibody that binds with high affinity and specificity to receptor activator of nuclear factor kappa-B ligand (RANKL). Denosumab blocks RANKL, preventing bone resorption and loss of bone density/architecture in conditions characterized by excessive bone loss such as osteoporosis in postmenopausal women and metastatic bone disease, among others. METHODS This narrative review summarizes the totality of evidence (ToE) for SDZ-deno that supported its approval as Jubbonti/Wyost in the EU and US. FINDINGS Analytical evaluation indicated that SDZ-deno has high purity and structural homology with REF-deno. SDZ-deno also demonstrated similar binding affinities, size and charge variants, and disulfide isoforms to REF-deno, and did not trigger clinically meaningful antibody-dependent cellular cytotoxicity. In clinical evaluation, SDZ-deno was similar to REF-deno in pharmacokinetics (PK) and pharmacodynamics (PD) in a 39-week Phase I study in 502 healthy male participants, and to REF-deno in a 72-week Phase III study in 527 postmenopausal women with osteoporosis. In both studies, the 90% and 95% confidence intervals (for PK and PD endpoints, respectively) of the geometric mean ratios for AUCinf, Cmax (and AUClast in the Phase I study; PK endpoints), and area under the effect versus time curve of percent change from baseline in serum carboxy-terminal crosslinked telopeptide of type I collagen (PD endpoint), were fully contained within the prespecified equivalence margins (0.80, 1.25). The Phase III study also demonstrated SDZ-deno is similar in efficacy to REF-deno in postmenopausal women with osteoporosis, as the difference in percent change from baseline in lumbar spine bone mineral density at week 52 between REF-deno and SDZ-deno was fully contained within the prespecified equivalence margins (-1.45, 1.45). SDZ-deno was well tolerated in both studies. As the ToE has established biosimilarity of SDZ-deno and REF-deno, extrapolation to all indications is justified based on the common mechanism of action and the comparable PK, safety, and immunogenicity across all indications. IMPLICATIONS The ToE for SDZ-deno suggests it will be an effective biosimilar to REF-deno, and its lower unit price is anticipated to increase the number of appropriate patients who will benefit.
Collapse
Affiliation(s)
- Barbara Vogg
- Hexal AG (a Sandoz company), Holzkirchen, Germany.
| | | | | | | | - Alan Kivitz
- Altoona Center for Clinical Research, Duncansville, Pennsylvania
| | | | | | - Jean-Jacques Body
- Department of Medicine, University Hospital Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Richard Eastell
- Division of Clinical Medicine, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
9
|
Dai HR, Guo HL, Hu YH, Liu Y, Lu KY, Zhang YY, Wang J, Ding XS, Jiao Z, Cheng R, Chen F. Development and application of a population pharmacokinetic model repository for caffeine dose tailoring in preterm infants. Expert Opin Drug Metab Toxicol 2024; 20:923-938. [PMID: 39167118 DOI: 10.1080/17425255.2024.2395561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/09/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND Considerable interindividual variability for the pharmacokinetics of caffeine in preterm infants has been demonstrated, emphasizing the importance of personalized dosing. This study aimed to develop and apply a repository of currently published population pharmacokinetic (PopPK) models of caffeine in preterm infants to facilitate model-informed precision dosing (MIPD). RESEARCH DESIGN AND METHODS Literature search was conducted using PubMed, Embase, Scopus, and Web of Science databases. Relevant publications were screened, and their quality was assessed. PopPK models were reestablished to develop the model repository. Covariate effects were evaluated and the concentration-time profiles were simulated. An online simulation and calculation tool was developed as an instance. RESULTS Twelve PopPK models were finally included in the repository. Preterm infants' age and body size, especially the postnatal age and current weight, were identified as the most clinically critical covariates. Simulated blood concentration-time profiles across these models were comparable. Caffeine citrate-dose regimen should be adjusted according to the age and body size of preterm infants. The developed online tool can be used to facilitate clinical decision-making. CONCLUSIONS The first developed repository of PopPK models for caffeine in preterm infants has a wide range of potential applications in the MIPD of caffeine.
Collapse
Affiliation(s)
- Hao-Ran Dai
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Li Guo
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Hu
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yun Liu
- Neonatal Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ke-Yu Lu
- Neonatal Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Zhang
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Wang
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuan-Sheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zheng Jiao
- Department of Pharmacy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Cheng
- Neonatal Intensive Care Unit, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Pharmaceutical Sciences Research Center, Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Jung D, Long-Boyle JR, Pang WW, Gobburu JVS. Pharmacokinetics of Briquilimab as a Conditioning Agent for Hematopoietic Stem Cell Transplantation in Patients With Severe Combined Immunodeficiency, Myelodysplastic Syndrome, or Acute Myeloid Leukemia. Transplant Cell Ther 2024; 30:923.e1-923.e9. [PMID: 38972509 DOI: 10.1016/j.jtct.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024]
Abstract
For successful engraftment of donor hematopoietic stem cells (HSC), conditioning with chemotherapy and/or radiation prior to hematopoietic cell transplantation (HCT) has been required to open marrow niche space and minimize the risk of immune rejection. Briquilimab, a humanized IgG1 monoclonal antibody that blocks the interaction between the c-Kit receptor and stem cell factor on various C-Kit expressing tissues including HSC, is a potential nonmyeloablative conditioning agent in clinical development for patients with severe combined immunodeficiency (SCID), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML). This study aimed to characterize pharmacokinetics (PK) and develop a population PK model of briquilimab after single intravenous infusions of 4 different doses in patients with SCID, MDS, or AML receiving HCT. The PK data was collected from 2 different studies: JAS-BMT-CP-001 and JSP-CP-003. JAS-BMT-CP-001 is a phase 1/2 open-label study of briquilimab as a conditioning agent prior to allogenic HCT in SCID patients. The participants received single intravenous infusions of 0.1, 0.3, 0.6, or 1.0 mg/kg. JSP-CP-003 was a phase 1a/b open-label study of briquilimab in combination with a standard conditioning regimen of low dose total body irradiation and fludarabine in MDS or AML subjects undergoing HCT. The participants received a single intravenous dose of 0.6 mg/kg briquilimab. In both studies, briquilimab PK samples were obtained at pre-treatment, 5 minutes post-end of infusion, 4- and 24-hours post-start of infusion, any time between 2 days and 30 days postinfusion, and on the day of HCT prior to donor cell infusion.The population PK model was developed using the PK data from these 2 clinical studies, and the effect of participants' baseline characteristics on the briquilimab PK was evaluated. PK simulations were performed using the developed PK model to calculate the time to reach target concentrations for HCT. A total of 49 participants (21 SCID adult and pediatric participants with a median age of 12 yr and 28 MDS/AML adult participants with a median age of 70 yr) were included in the PK analysis. A 2-compartment model with combined linear and non-linear elimination best described the PK of briquilimab. Body weight was determined as the sole covariate of the PK parameters among the explored covariates. For a typical subject with a body weight 70 kg, the estimated parameters for clearance, maximum metabolic rate of Michaelis Menten elimination, Michaelis Menten constant, central volume, peripheral volume, and intercompartmental clearance were 17.6 mL/h, 51,434.8 ng/h, 71.5 ng/mL, 3444.0 mL, 1613.3 mL, and 21.2 mL/h, respectively. The median time to reach target concentrations of 500, 1000, and 2000 ng/mL after a single dose of 0.6 mg/kg was calculated as 12.3, 10.4, and 7.7 days, respectively. The PK of intravenous briquilimab was characterized in subjects with SCID, MDS, or AML receiving HCT, and a population PK model was developed to estimate briquilimab clearance to use as a guide to the timing of donor cell infusion post-briquilimab. Body weight was identified as a significant covariate on elimination and volume of distribution of briquilimab.
Collapse
Affiliation(s)
- Dawoon Jung
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Janel R Long-Boyle
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Wendy W Pang
- Jasper Therapeutics Inc., Redwood City, California
| | - Jogarao V S Gobburu
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland.
| |
Collapse
|
11
|
Zhang D, Zhang Z, Lee A, Fenton K, Jain S, Garg A, Chia YL. Time-varying brentuximab vedotin pharmacokinetics and weight-based dosing in paediatric patients despite lower exposure in those aged 2 to <6 and 6-11 years. Br J Clin Pharmacol 2024; 90:2299-2313. [PMID: 38866401 DOI: 10.1111/bcp.16128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 06/14/2024] Open
Abstract
AIMS We studied the pharmacokinetics and exposure-response relationships of the brentuximab vedotin (BV) antibody-drug conjugate (ADC) and unconjugated monomethyl auristatin E in haematologic malignancies. METHODS This population pharmacokinetic analysis included data from five adult and three paediatric studies. Exposures in virtual adult and paediatric populations following BV 1.8 mg/kg (maximum 180 mg) intravenously every 3 weeks were simulated. Clinical endpoints included overall response rate, grade ≥2 peripheral neuropathy (PN) and grade ≥3 neutropenia. RESULTS BV ADC exhibited linear pharmacokinetics, well-described by a three-compartment model, with body weight being the only significant covariate for exposure. Monomethyl auristatin E exhibited time-varying formation rate. Simulated steady-state BV ADC exposures in patients aged 12 to <18 years were similar to those of adult patients, but 23%-38% lower in patients aged 2 to <12 years. Despite lower exposure, clinical activity was observed with BV 1.8 mg/kg every 3 weeks in those aged 2 to <12 years (overall response rate: 2 to <12 years, 60%; 12 to <18 years, 43%). In adult, but not paediatric patients, increased BV ADC exposures were associated with grade ≥2 PN and grade ≥3 neutropenia occurrence. CONCLUSIONS BV pharmacokinetics in adult and paediatric patients were consistent. BV ADC exposures were lower in patients aged 2 to <12 years vs. ≥12 years, but no apparent clinically relevant differences in efficacy, grade ≥2 PN or grade ≥3 neutropenia were observed. These data support body weight-based dosing of BV in patients irrespective of age; thus, dose adjustment in those 2 to <12 years does not appear warranted.
Collapse
Affiliation(s)
- Daping Zhang
- Translational Clinical Sciences, Research and Development, Pfizer, Bothell, Washington, USA
| | - Zufei Zhang
- Clinical Pharmacology and Translational Sciences, Oncology Research and Development, Pfizer, Bothell, Washington, USA
| | - Anthony Lee
- Clinical Pharmacology and Translational Sciences, Oncology Research and Development, Pfizer, Bothell, Washington, USA
| | - Keenan Fenton
- Oncology Statistics, Oncology Research and Development, Pfizer, Bothell, Washington, USA
| | - Shweta Jain
- Oncology Research and Development, Pfizer, Bothell, Washington, USA
| | - Amit Garg
- Clinical Pharmacology and Translational Sciences, Oncology Research and Development, Pfizer, South San Francisco, California, USA
| | - Yen Lin Chia
- Translational Clinical Sciences, Research and Development, Pfizer, South San Francisco, California, USA
| |
Collapse
|
12
|
Zhao Y, Hu Z, Bathena SP, Keidel S, Miller-Moslin K, Statkevich P, Bello A, Roy A, Suryawanshi S. Model-Informed Clinical Pharmacology Profile of a Novel Fixed-Dose Combination of Nivolumab and Relatlimab in Adult and Adolescent Patients with Solid Tumors. Clin Cancer Res 2024; 30:3050-3058. [PMID: 38295151 PMCID: PMC11247312 DOI: 10.1158/1078-0432.ccr-23-2396] [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: 08/11/2023] [Revised: 10/27/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE Progression-free survival (PFS) was significantly improved with nivolumab 480 mg plus relatlimab 160 mg fixed-dose combination (FDC) every 4 weeks (Q4W) versus nivolumab alone in patients with previously untreated advanced melanoma in RELATIVITY-047. In addition, RELATIVITY-020 (Part D) demonstrated a manageable safety profile and potential for durable response with nivolumab plus relatlimab in previously treated patients. Here, we evaluate the clinical pharmacology profile (CPP) of nivolumab plus relatlimab to support the approved regimen for adult and adolescent patients with advanced melanoma and its continued clinical development in solid tumors. EXPERIMENTAL DESIGN The pharmacokinetics (PK) and immunogenicity of relatlimab and nivolumab were assessed using data from RELATIVITY-047 and RELATIVITY-020. Patients with advanced solid tumors received relatlimab alone or nivolumab plus relatlimab as single-agent vials (SAV) or FDC. PK was characterized using a population PK (popPK) model. RESULTS Relatlimab demonstrated nonlinear and time-varying PK. Nonlinearity in relatlimab PK represented approximately 31% of total CL of relatlimab 160 mg Q4W. Relatlimab PK was dose proportional at doses ≥160 mg Q4W. Geometric mean exposures were similar for SAV and FDC cohorts receiving equivalent dosing regimens. No dose adjustment was required for covariates. Incidence of relatlimab antidrug antibodies was <6% for nivolumab plus relatlimab and had no clinically meaningful impact. There was no PK-related drug interaction of nivolumab plus relatlimab. CONCLUSIONS The CPP of relatlimab alone or in combination with nivolumab supports the approved dosing in advanced melanoma and the continued evaluation of nivolumab and relatlimab across other solid tumors. See related commentary by Gopalakrishnan and Amaria, p. 2862.
Collapse
Affiliation(s)
- Yue Zhao
- Bristol-Myers Squibb, Princeton, New Jersey
| | - Zheyi Hu
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | | | | | | | | | - Amit Roy
- Bristol-Myers Squibb, Princeton, New Jersey
| | | |
Collapse
|
13
|
Leek LVM, Notohardjo JCL, de Joode K, Velker EL, Haanen JBAG, Suijkerbuijk KPM, Aarts MJB, de Groot JWB, Kapiteijn E, van den Berkmortel FWPJ, Westgeest HM, de Gruijl TD, Retel VP, Cuppen E, van der Veldt AAM, Labots M, Voest EE, van de Haar J, van den Eertwegh AJM. Multi-omic analysis identifies hypoalbuminemia as independent biomarker of poor outcome upon PD-1 blockade in metastatic melanoma. Sci Rep 2024; 14:11244. [PMID: 38755213 PMCID: PMC11099084 DOI: 10.1038/s41598-024-61150-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: 02/09/2024] [Accepted: 05/02/2024] [Indexed: 05/18/2024] Open
Abstract
We evaluated the prognostic value of hypoalbuminemia in context of various biomarkers at baseline, including clinical, genomic, transcriptomic, and blood-based markers, in patients with metastatic melanoma treated with anti-PD-1 monotherapy or anti-PD-1/anti-CTLA-4 combination therapy (n = 178). An independent validation cohort (n = 79) was used to validate the performance of hypoalbuminemia compared to serum LDH (lactate dehydrogenase) levels. Pre-treatment hypoalbuminemia emerged as the strongest predictor of poor outcome for both OS (HR = 4.01, 95% CI 2.10-7.67, Cox P = 2.63e-05) and PFS (HR = 3.72, 95% CI 2.06-6.73, Cox P = 1.38e-05) in univariate analysis. In multivariate analysis, the association of hypoalbuminemia with PFS was independent of serum LDH, IFN-γ signature expression, TMB, age, ECOG PS, treatment line, treatment type (combination or monotherapy), brain and liver metastasis (HR = 2.76, 95% CI 1.24-6.13, Cox P = 0.0131). Our validation cohort confirmed the prognostic power of hypoalbuminemia for OS (HR = 1.98, 95% CI 1.16-3.38; Cox P = 0.0127) and was complementary to serum LDH in analyses for both OS (LDH-adjusted HR = 2.12, 95% CI 1.2-3.72, Cox P = 0.00925) and PFS (LDH-adjusted HR = 1.91, 95% CI 1.08-3.38, Cox P = 0.0261). In conclusion, pretreatment hypoalbuminemia was a powerful predictor of outcome in ICI in melanoma and showed remarkable complementarity to previously established biomarkers, including high LDH.
Collapse
Affiliation(s)
- Lindsay V M Leek
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jessica C L Notohardjo
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Karlijn de Joode
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Eline L Velker
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, UMC Utrecht Cancer Center, Utrecht University, Utrecht, The Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan Willem B de Groot
- Department of Medical Oncology, Oncology Center Isala, Isala, Zwolle, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Hans M Westgeest
- Department of Medical Oncology, Amphia Hospital, Breda, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Valesca P Retel
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
- Health Technology and Services Research Department, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Edwin Cuppen
- Hartwig Medical Foundation, Amsterdam, The Netherlands
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Mariette Labots
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Emile E Voest
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Joris van de Haar
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, The Netherlands.
| | | |
Collapse
|
14
|
Suzuki S, Taguchi Y, Kitabayashi T, Sato N, Kaya H, Abe T, Endo T, Suzuki H, Kawasaki Y, Yamada T. Serum Albumin as an Independent Predictor of Long-Term Survival in Patients with Recurrent and Metastatic Head and Neck Squamous Cell Carcinoma Treated with Nivolumab. J Clin Med 2024; 13:2456. [PMID: 38730986 PMCID: PMC11084251 DOI: 10.3390/jcm13092456] [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: 04/01/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Nivolumab has been shown to improve the overall survival (OS) of patients with recurrent and metastatic head and neck squamous cell carcinoma (R/M HNSCC). However, there is a need to identify factors associated with long-term survival (beyond 2 years) in these patients. This study investigated the relationship between pretreatment factors and long-term survival in patients with R/M HNSCC treated with nivolumab. Methods: Forty-nine patients with R/M HNSCC who were treated with nivolumab were retrospectively reviewed. Baseline characteristics, clinical data, and survival outcomes were evaluated. Univariate and multivariate analyses were performed to identify factors associated with long-term survival (OS ≥ 2 years). Results: The median OS in the overall cohort was 11.0 months, and the 2-year survival rate was 34.7%. Long-term survivors (OS ≥ 2 years) had significantly higher proportions of patients with Eastern Cooperative Oncology Group (ECOG) performance status (PS) scores of 0 or 1, serum albumin levels ≥ 3.5 g/dL, and neutrophil-to-eosinophil ratio (NER) < 32.0 compared to non-long-term survivors. On multivariate analysis, serum albumin levels ≥ 3.5 g/dL, in addition to ECOG-PS score of 0 or 1, were independent predictors of long-term survival. Conclusions: Pretreatment serum albumin levels may be useful for predicting long-term survival in R/M HNSCC patients treated with nivolumab.
Collapse
Affiliation(s)
- Shinsuke Suzuki
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Makarenko I, Petrov A, Belova B, Saparova V, Arefeva A, Peskov K, Kudryashova N, Khokhlov A, Drai R. Population Pharmacokinetic and Pharmacodynamic Modeling of Romiplostim Biosimilar GP40141 and Reference Product in Healthy Volunteers to Evaluate Biosimilarity. Clin Pharmacol Drug Dev 2024; 13:419-431. [PMID: 38168134 DOI: 10.1002/cpdd.1367] [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: 08/31/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
GP40141 is a romiplostim biosimilar. A Phase 1 clinical trial was previously conducted in healthy volunteers to evaluate the pharmacodynamics (PD), pharmacokinetics (PK), and safety of GP40141 compared to the reference romiplostim (NCT05652595). Using noncompartmental analysis, the biosimilarity of PD end points was determined according to the classical criterion (0.8-1.25). PK end points were also in good agreement between GP40141 and the reference romiplostim; however, the confidence interval for the area under concentration-time curve from time 0 to the time of last measurement was slightly out of the bioequivalence range (0.91-1.29). Population PK/PD was used in the present study to characterize the individual PK and PD data of 56 healthy subjects in 2 cross-over periods of the Phase 1 clinical trial. Body weight and neutralizing antibodies to romiplostim were found to be important predictors of apparent volume of distribution and linear elimination constant, respectively. Within the framework of the conducted modeling, population estimates of PK/PD parameters were obtained, which were in agreement with literature data for the reference romiplostim. Additionally, values of intersubject variability, previously unreported for romiplostim in a healthy subject population, were derived. Covariate analysis, conducted during model development, as well as visual diagnostics and model-based simulations, demonstrated the absence of significant differences in PK and PD between GP40141 and romiplostim-ref.
Collapse
Affiliation(s)
| | | | | | | | | | - Kirill Peskov
- Modeling and Simulation Decisions FZ - LLC, Dubai, United Arab Emirates
- Sirius University of Science and Technology, Sirius, Russia
- Research Center of Model-Informed Drug Development, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nataliya Kudryashova
- Research Center of Model-Informed Drug Development, Sechenov First Moscow State Medical University, Moscow, Russia
- V.L. Talrose Institute for Energy Problems of Chemical Physics of Russian Academy of Science, Moscow, Russia
- Semenov Research Center of Chemical Physics, Moscow, Russia
| | - Alexandr Khokhlov
- Federal State Budgetary Educational Institution of Higher Education, "Yaroslavl State Medical University" of the Ministry of Health of the Russian Federation, Yaroslavl, Russia
| | - Roman Drai
- R&D Center, GEROPHARM, Saint-Petersburg, Russia
| |
Collapse
|
16
|
Hsu JC, Wu M, Kim C, Vora B, Lien YTK, Jindal A, Yoshida K, Kawakatsu S, Gore J, Jin JY, Lu C, Chen B, Wu B. Applications of Advanced Natural Language Processing for Clinical Pharmacology. Clin Pharmacol Ther 2024; 115:786-794. [PMID: 38140747 DOI: 10.1002/cpt.3161] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/20/2023] [Indexed: 12/24/2023]
Abstract
Natural language processing (NLP) is a branch of artificial intelligence, which combines computational linguistics, machine learning, and deep learning models to process human language. Although there is a surge in NLP usage across various industries in recent years, NLP has not been widely evaluated and utilized to support drug development. To demonstrate how advanced NLP can expedite the extraction and analyses of information to help address clinical pharmacology questions, inform clinical trial designs, and support drug development, three use cases are described in this article: (1) dose optimization strategy in oncology, (2) common covariates on pharmacokinetic (PK) parameters in oncology, and (3) physiologically-based PK (PBPK) analyses for regulatory review and product label. The NLP workflow includes (1) preparation of source files, (2) NLP model building, and (3) automation of data extraction. The Clinical Pharmacology and Biopharmaceutics Summary Basis of Approval (SBA) documents, US package inserts (USPI), and approval letters from the US Food and Drug Administration (FDA) were used as our source data. As demonstrated in the three example use cases, advanced NLP can expedite the extraction and analyses of large amounts of information from regulatory review documents to help address important clinical pharmacology questions. Although this has not been adopted widely, integrating advanced NLP into the clinical pharmacology workflow can increase efficiency in extracting impactful information to advance drug development.
Collapse
Affiliation(s)
- Joy C Hsu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Michael Wu
- Computational Sciences, Genentech, Inc., South San Francisco, California, USA
| | - Chloe Kim
- Computational Sciences, Genentech, Inc., South San Francisco, California, USA
| | - Bianca Vora
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Yi Ting Kayla Lien
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Ashutosh Jindal
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Kenta Yoshida
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Sonoko Kawakatsu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
- A2-Ai, Ann Arbor, Michigan, USA
| | - Jeremy Gore
- Capgemini America, Inc., New York, New York, USA
| | - Jin Y Jin
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Christina Lu
- Computational Sciences, Genentech, Inc., South San Francisco, California, USA
| | - Bingyuan Chen
- Computational Sciences, Genentech, Inc., South San Francisco, California, USA
| | - Benjamin Wu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
17
|
Haddish-Berhane N, Su Y, Russu A, Thayu M, Knoblauch RE, Mehta J, Xie J, Gibbs E, Sun YN, Zhou H. Determination and Confirmation of Recommended Ph2 Dose of Amivantamab in Epidermal Growth Factor Receptor Exon 20 Insertion Non-Small Cell Lung Cancer. Clin Pharmacol Ther 2024; 115:468-477. [PMID: 37776107 DOI: 10.1002/cpt.3064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/10/2023] [Indexed: 10/01/2023]
Abstract
Amivantamab has demonstrated durable responses with a tolerable safety profile in non-small cell lung cancer with EGFR exon 20 insertions (Ex20ins) who progressed after prior platinum chemotherapy. Data supporting the amivantamab recommended phase II dose (RP2D) in this patient population are presented. Pharmacokinetic (PK) analysis and population PK (PopPK) modeling were conducted using serum concentration data obtained following amivantamab intravenous administration (140-1,750 mg). Pharmacodynamics (PDs) were evaluated using depletion of soluble EGFR and MET. Exposure-response (E-R) analyses were performed using the primary efficacy end point of objective response rate in patients with EGFR Ex20ins. The E-R relationship for safety was explored for adverse events of clinical interest. Amivantamab exhibited linear PKs at 350-1,750 mg dose levels following administration, with no maximum tolerated dose identified. A two-compartment PopPK model with linear clearance adequately described the observed PKs. Body weight was a covariate of clearance and volume of distribution in the central compartment. PopPK modeling showed that a weight-based, 2-tier (< 80 and ≥ 80 kg) dosing strategy reduces PK variability and provides comparable exposure across 2 weight groups, with 87% of patients achieving exposures above the target threshold. The final confirmed RP2D of amivantamab was 1,050 mg for < 80 kg (1,400 mg for ≥ 80 kg) weekly in cycle 1 (28 days) and every 2 weeks thereafter. No significant exposure-efficacy or safety correlation was observed. In conclusion, the amivantamab RP2D is supported by PK, PD, safety, and efficacy analyses. E-R analyses confirmed that the current regimen provides durable efficacy with tolerable safety.
Collapse
Affiliation(s)
| | - Yaming Su
- Janssen Research & Development, LLC, Raritan, New Jersey, USA
| | - Alberto Russu
- Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Meena Thayu
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | | | - Jaydeep Mehta
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - John Xie
- Janssen Research & Development, LLC, Raritan, New Jersey, USA
| | - Eric Gibbs
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| | - Yu-Nien Sun
- Cognigen Division, Simulations-Plus Company, Buffalo, New York, USA
| | - Honghui Zhou
- Jazz Pharmaceuticals, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Guo Y, Quijano Cardé NA, Kang L, Verona R, Banerjee A, Kobos R, Chastain K, Uhlar C, Pillarisetti K, Doyle M, Smit J, Haddish‐Berhane N, Ouellet D. Teclistamab: Mechanism of action, clinical, and translational science. Clin Transl Sci 2024; 17:e13717. [PMID: 38266057 PMCID: PMC10784707 DOI: 10.1111/cts.13717] [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: 08/18/2023] [Revised: 10/31/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Multiple myeloma (MM) remains incurable despite improvements in treatment options. B-cell maturation antigen (BCMA) is predominantly expressed in B-lineage cells and represents a promising new target for MM. Teclistamab (TECVAYLITM ) is the first T-cell redirecting bispecific antibody approved for patients with MM. Targeting both CD3 receptor complex on T cells and BCMA on myeloma cells, teclistamab leads to T-cell activation and subsequent lysis of BCMA+ cells. The recommended dose of teclistamab is 1.5 mg/kg subcutaneous weekly after two step-up doses of 0.06 and 0.3 mg/kg, which was selected after review of safety, efficacy, pharmacokinetic, and pharmacodynamic data. Exposure-response analyses of efficacy and safety data were also used to confirm the teclistamab dose. Teclistamab resulted in a high rate of deep and durable responses (63% overall response, 45.5% complete response or better, with 22 months median duration of response) in patients with triple-exposed relapsed/refractory MM. Common adverse reactions included cytokine release syndrome, hematologic abnormalities, and infections. Teclistamab is currently being investigated as monotherapy as well as combination therapy across different MM indications.
Collapse
Affiliation(s)
- Yue Guo
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | - Lijuan Kang
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Raluca Verona
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Arnob Banerjee
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Rachel Kobos
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | - Clarissa Uhlar
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | | | - Jennifer Smit
- Janssen Research & DevelopmentSpring HousePennsylvaniaUSA
| | | | | |
Collapse
|
19
|
Proctor JR, Wong H. Time-dependent clearance can confound exposure-response analysis of therapeutic antibodies: A comprehensive review of the current literature. Clin Transl Sci 2024; 17:e13676. [PMID: 37905360 PMCID: PMC10766027 DOI: 10.1111/cts.13676] [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: 09/01/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Exposure-response (ER) analysis is used to optimize dose and dose regimens during clinical development. Characterization of relationships between drug exposure and efficacy or safety outcomes can be utilized to make dose adjustments that improve patient response. Therapeutic antibodies typically show predictable pharmacokinetics (PK) but can exhibit clearance that decreases over time due to treatment. Moreover, time-dependent changes in clearance are frequently associated with drug response, with larger decreases in clearance and increased exposure seen in patients who respond to treatment. This often confounds traditional ER analysis, as drug response influences exposure rather than the reverse. In this review, we survey published population PK analyses for reported time-dependent drug clearance effects across 158 therapeutic antibodies approved or in regulatory review. We describe the mechanisms by which time-dependent clearance can arise, and evaluate trends in frequency, magnitude, and time scale of changes in clearance with respect to indication, mechanistic interpretation of time-dependence, and PK modeling techniques employed. We discuss the modeling and simulation strategies commonly used to characterize time-dependent clearance, and examples where time-dependent clearance has impeded ER analysis. A case study using population model simulation was explored to interrogate the impact of time-dependent clearance on ER analysis and how it can lead to spurious conclusions. Overall, time-dependent clearance arises frequently among therapeutic antibodies and has spurred erroneous conclusions in ER analysis. Appropriate PK modeling techniques aid in identifying and characterizing temporal shifts in exposure that may impede accurate ER assessment and successful dose optimization.
Collapse
Affiliation(s)
- Jeffrey R. Proctor
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Harvey Wong
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
20
|
Zhang N, Chan ML, Li J, Brohawn PZ, Sun B, Vainshtein I, Roskos LK, Faggioni R, Savic RM. Combining pharmacometric models with predictive and prognostic biomarkers for precision therapy in Crohn's disease: A case study of brazikumab. CPT Pharmacometrics Syst Pharmacol 2023; 12:1945-1959. [PMID: 37691451 PMCID: PMC10725267 DOI: 10.1002/psp4.13044] [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: 04/08/2023] [Revised: 06/11/2023] [Accepted: 07/11/2023] [Indexed: 09/12/2023] Open
Abstract
Pharmacometric models were used to investigate the utility of biomarkers in predicting the efficacy (Crohn's Disease Activity Index [CDAI]) of brazikumab and provide a data-driven framework for precision therapy for Crohn's disease (CD). In a phase IIa trial in patients with moderate to severe CD, treatment with brazikumab, an anti-interleukin 23 monoclonal antibody, was associated with clinical improvement. Brazikumab treatment effect was determined to be dependent on the baseline IL-22 (BIL22) or baseline C-reactive protein (BCRP; predictive biomarkers), and placebo effect was found to be correlated with the baseline CDAI (a prognostic biomarker). A maximal total inhibition on CDAI input function of 50.6% and 42.4% was predicted for patients with extremely high BIL22 or BCRP, compared to a maximal total inhibition of 20.9% and 17.8% for patients with extremely low BIL22 or BCRP, respectively, which were mainly due to the placebo effect. We demonstrated that model-derived baseline biomarker levels that achieve 50% of maximum unbound systemic concentration of 22.8 pg/mL and 8.03 mg/L for BIL22 and BCRP as the cutoffs to select subpopulations can effectively identify high-response subgroup patients with improved separation of responders when compared to using the median values as the cutoff. This work exemplifies the utility of pharmacometrics to quantify biomarker-driven responses in biologic therapies and distinguish between predictive and prognostic biomarkers, complementing clinical efforts of identifying subpopulations with higher likelihood of response to brazikumab.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ming Liang Chan
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jing Li
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Philip Z. Brohawn
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceutical R&D, AstraZenecaGaithersburgMarylandUSA
| | - Bo Sun
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Inna Vainshtein
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Lorin K. Roskos
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Raffaella Faggioni
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences (CPSS), R&D, AstraZenecaSouth San FranciscoCaliforniaUSA
| | - Rada M. Savic
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
21
|
Sun Y, Li C, Wang X, Zheng Y, Wu Z, Hui AM, Diao L. Model-informed dose selection for an investigational human epidermal growth factor receptor 2 antibody-drug conjugate FS-1502 in patients with human epidermal growth factor receptor 2-expressing advanced malignant solid tumours. Br J Clin Pharmacol 2023. [PMID: 37926561 DOI: 10.1111/bcp.15955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
AIMS The dose-escalation phase (phase Ia study) of a novel human epidermal growth factor receptor 2 (HER2) antibody-drug conjugate (ADC) FS-1502 included a dose range from 0.1 to 3.5 mg/kg in HER2-expressing advanced malignant solid tumours. However, the defined maximum tolerated dose was not reached. This model-informed approach integrated population pharmacokinetic (PopPK) modelling and exposure-response (E-R) analysis to facilitate dose selection for phase II. METHODS The PopPK model was constructed using PK data from 109 Chinese patients who received doses of 0.1-3.5 mg/kg FS-1502 every 3 (Q3W) or 4 weeks during a phase I dose-escalation and dose expansion trial. The structural model consisted of compartment models for FS-1502 and unconjugated monomethyl auristatin F. E-R was explored for the percentage change in tumour size, overall response rate and treatment-related adverse events. RESULTS A semi-mechanistic 2-analyte PopPK model was developed. The FS-1502 PK data were best described by a 2-compartment PK model with parallel linear and nonlinear Michaelis-Menten eliminations. The PK of unconjugated monomethyl auristatin F was described by a 2-compartment model with first-order elimination. E-R analysis supported the clinically meaningful efficacy of FS-1502 at 2.3 mg/kg and above. However, 2.3 mg/kg Q3W was considered to have a better benefit-risk balance due to a lower incidence of safety events without a significant reduction in efficacy compared to 3.0 mg/kg Q3W. CONCLUSION This PopPK and E-R analysis guided the recommended phase II dose selection of 2.3 mg/kg Q3W and supported body weight-based dosing for an investigational HER2 ADC FS-1502.
Collapse
Affiliation(s)
- Yi Sun
- Shanghai Fosun Pharmaceutical Development Co., Ltd., Shanghai, China
| | - Chao Li
- Anheart Therapeutics, New York, New York, USA
| | - Xingli Wang
- Shanghai Fosun Pharmaceutical Development Co., Ltd., Shanghai, China
| | - Yi Zheng
- Shanghai Fosun Pharmaceutical Development Co., Ltd., Shanghai, China
| | - Zhuli Wu
- Shanghai Fosun Pharmaceutical Development Co., Ltd., Shanghai, China
| | | | - Lei Diao
- Shanghai Fosun Pharmaceutical Development Co., Ltd., Shanghai, China
| |
Collapse
|
22
|
Yoshihara K, Kobayashi Y, Endo S, Fukae M, Hennig S, Kastrissios H, Kamiyama E, Garimella T, Abutarif M. Trastuzumab Deruxtecan Dosing in Human Epidermal Growth Factor Receptor 2-Positive Gastric Cancer: Population Pharmacokinetic Modeling and Exposure-Response Analysis. J Clin Pharmacol 2023; 63:1232-1243. [PMID: 37393579 DOI: 10.1002/jcph.2295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023]
Abstract
This study evaluated the benefit/risk of trastuzumab deruxtecan (T-DXd) 6.4 mg/kg in patients with human epidermal growth factor receptor 2 (HER2)-positive gastric cancer using pharmacometrics. A population pharmacokinetic (PopPK) model was developed using data from patients with gastric cancer, breast cancer, or other tumors in T-DXd clinical trials, primarily conducted in Asia. Post hoc model-estimated pharmacokinetic metrics were used in exposure-efficacy (objective response rates, ORRs) and exposure-safety analyses. The PopPK analysis included 808 patients (217 with gastric cancer, 512 with breast cancer, and 79 with other cancers). In gastric cancer, the T-DXd 6.4 mg/kg steady-state exposure metrics were lower compared with 6.4 mg/kg in breast cancer, but were similar to 5.4 mg/kg in breast cancer. Tumor type was selected as a significant covariate on T-DXd clearance. In exposure-efficacy analysis among 160 patients with gastric cancer, the T-DXd steady-state minimum concentration was associated with a confirmed ORR in univariate logistic regression analysis (P = .023). The model-predicted confirmed ORRs in gastric cancer were 36.0% (90%CI 29.3% to 43.7%) with 5.4 mg/kg and 40.0% (90%CI 33.1% to 47.6%) with 6.4 mg/kg. Among 808 patients in the exposure-safety analyses, the model-predicted estimates for the rates of any-grade interstitial lung disease (ILD) over a period of 180 days were 10.2% (90%CI 8.7% to 12.8%) with 6.4 mg/kg in gastric cancer and 9.7% (90%CI 8.2% to 11.8%) with 5.4 mg/kg in breast cancer. In gastric cancer, the efficacy of T-DXd was higher at 6.4 mg/kg than at 5.4 mg/kg. Exposure and ILD rates were comparable between 6.4 mg/kg in gastric cancer and 5.4 mg/kg in breast cancer. This study identified T-DXd 6.4 mg/kg as the recommended dose in HER2-positive gastric cancer.
Collapse
Affiliation(s)
- Kazutaka Yoshihara
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Seiko Endo
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Masato Fukae
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Helen Kastrissios
- Certara Inc., Melbourne, Australia
- QuanTx Consulting, Mountain View, CA, USA
| | - Emi Kamiyama
- Quantitative Clinical Pharmacology, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Tushar Garimella
- Quantitative Clinical Pharmacology, Daiichi Sankyo Inc., Basking Ridge, NJ, USA
| | - Malaz Abutarif
- Quantitative Clinical Pharmacology, Daiichi Sankyo Inc., Basking Ridge, NJ, USA
| |
Collapse
|
23
|
Chua L, Friedrich S, Zhang XC. Mirikizumab Pharmacokinetics in Patients with Moderately to Severely Active Ulcerative Colitis: Results from Phase III LUCENT Studies. Clin Pharmacokinet 2023; 62:1479-1491. [PMID: 37610533 PMCID: PMC10520102 DOI: 10.1007/s40262-023-01281-z] [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] [Accepted: 06/22/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND AND OBJECTIVE Mirikizumab is a humanized anti-interleukin-23-p19 monoclonal antibody being developed for ulcerative colitis and Crohn's disease. This analysis characterized mirikizumab pharmacokinetics using phase II and III trial data from patients with moderately to severely active ulcerative colitis. METHODS Serum pharmacokinetic data in patients receiving mirikizumab 50-1000 mg intravenously every 4 weeks as induction treatment and mirikizumab 200 mg subcutaneously every 4 or 12 weeks as maintenance treatment across three trials (N = 1362) were analyzed using non-linear mixed-effects modeling. Covariate effects on mirikizumab exposure were evaluated using simulation-based estimations. RESULTS Mirikizumab pharmacokinetics was best described by a linear two-compartment model with first-order absorption. Clearance, volume of distribution for central and peripheral compartments, and half-life were estimated at approximately 0.022 L/h (linear), 3.11 L and 1.69 L, and 9.5 days, respectively. Statistically significant effects of body weight and serum albumin levels on clearance, body weight on central and peripheral volumes of distribution, and body mass index on bioavailability were observed but effects were small relative to random inter-individual variability (% coefficient of variation: 18-64%). The subcutaneous bioavailability of mirikizumab was 48%. CONCLUSIONS Mirikizumab displayed pharmacokinetic characteristics typical of a monoclonal antibody where clearance increased with body weight and decreased with the albumin level, and bioavailability decreased with body mass index. These effects were small relative to random variability, indicating that a dose adjustment for patient factors is not required. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov: NCT02589665 (28 October, 2015), NCT03518086 (8 May, 2018), NCT03524092 (14 May, 2018).
Collapse
Affiliation(s)
- Laiyi Chua
- Global PK/PD and Pharmacometrics, Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN, 46225, USA
| | - Stuart Friedrich
- Global PK/PD and Pharmacometrics, Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN, 46225, USA
| | - Xin Cindy Zhang
- Global PK/PD and Pharmacometrics, Eli Lilly and Company, 893 Delaware Street, Indianapolis, IN, 46225, USA.
| |
Collapse
|
24
|
Chen L, Wu F, Chen X, Chen Y, Deng L, Cai Q, Wu L, Guo W, Chen M, Li Y, Zhang W, Jin X, Chen H, Nie Q, Wu X, Lin Y, Wang C, Fu F. Impact of body mass index in therapeutic response for HER2 positive breast cancer treated with neoadjuvant targeted therapy: a multi-center study and meta-analysis. NPJ Breast Cancer 2023; 9:46. [PMID: 37258524 DOI: 10.1038/s41523-023-00552-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 05/19/2023] [Indexed: 06/02/2023] Open
Abstract
While overweight/obesity has become a major public health issue worldwide, any association between body mass index (BMI) and therapeutic response in neoadjuvant targeted therapy treated HER2 positive breast cancer patients remain unclear. The information from a total of four-hundred and ninety-one neoadjuvant targeted therapy treated HER2 positive breast cancer patients from four institutions were retrospectively collected. Univariate and multivariate logistic analysis was developed to determine the association between BMI and therapeutic response. A meta-analysis of published literature was then conducted to confirm the effect of overweight/obesity on pCR for patients treated with neoadjuvant targeted therapy. Restricted cubic spline (RCS) adjusted for confounding factors demonstrated a decrease pCR with increasing BMI (OR = 0.937, P = 0.045). Patients were then categorized into under/normal weight (n = 299) and overweight/obesity (n = 192). Overweight/obese patients were independently associated with a poor therapeutic response. In the subgroup analysis, a significant negative impact of overweight/obesity on pCR can be observed both in single-targeted (OR = 0.556; P = 0.02) and dual-targeted (OR = 0.392; P = 0.021) populations. Six eligible studies involving 984 neoadjuvant targeted therapy treated HER2 positive breast cancer patients were included in the meta-analysis. The meta-analysis also demonstrated that overweight/obesity was significantly associated with a poor response to neoadjuvant anti-HER2 therapy (OR = 0.68; P = 0.007). Our result show that overweight and obese HER2 positive breast cancer patients are less likely to achieve pCR after neoadjuvant targeted therapy.
Collapse
Affiliation(s)
- Lili Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Fan Wu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, People's Republic of China
| | - Xiaobin Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Yazhen Chen
- Department of Breast Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 363000, Zhangzhou, Fujian, P.R. China
| | - Lin Deng
- Department of Oncology, No. 900 Hospital of The Joint Logistic Support Force, Fuzhou, China
| | - Qindong Cai
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Long Wu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wenhui Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Minyan Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Yan Li
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Wenzhe Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Xuan Jin
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Hanxi Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Qian Nie
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China
| | - Xiong Wu
- Department of Breast Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, 363000, Zhangzhou, Fujian, P.R. China.
| | - Yuxiang Lin
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China.
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China.
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China.
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China.
| | - Fangmeng Fu
- Department of Breast Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China.
- Department of General Surgery, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian Province, China.
- Breast Cancer Institute, Fujian Medical University, 350001, Fuzhou, Fujian Province, China.
| |
Collapse
|
25
|
de Vries F, Smit AAJ, Wolbink G, de Vries A, Loeff FC, Franssen EJF. Case report: Pharmacokinetics of pembrolizumab in a patient with stage IV non-small cell lung cancer after a single 200 mg administration. Front Oncol 2023; 12:960116. [PMID: 36713570 PMCID: PMC9875126 DOI: 10.3389/fonc.2022.960116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/16/2022] [Indexed: 01/13/2023] Open
Abstract
Background Pembrolizumab is a well-tolerated biologic agent with a potentially stable and durable anti-tumor response. Unfortunately, discontinuation of therapy can occur as a consequence of immune-related adverse effects (irAEs). These irAEs appear independent of dose and exposure. However, such irAEs might also result from pembrolizumab's highly specific mechanism of action and current dosing regimens. However, the currently available pharmacokinetic (PK) and pharmacodynamic (PD) data to reassess dosing strategies are insufficient.To highlight the importance of additional PK/PD studies, we present a case describing the complexity of pembrolizumab's PK/PD after a single 200 mg pembrolizumab dose in a treatment-naive patient with non-small cell lung cancer (NSCLC). Case description A 72-year-old man with stage IV NSCLC presented hepatotoxic symptoms 19 days after receiving the first 200 mg pembrolizumab dose. Hence, pembrolizumab therapy was paused, and prednisolone therapy was initiated, which successfully inhibited the toxic effect of pembrolizumab. However, repeated flare-ups due to prednisolone tapering suggest that the toxic effect of pembrolizumab outlasts the presence of pembrolizumab in the bloodstream. This further suggests that the T-cell-mediated immune response outlasts the programmed cell death protein 1 (PD-1) receptor occupancy by pembrolizumab, which challenges the need for the current fixed-interval strategies and their stop criteria.Furthermore, a validated ELISA quantified pembrolizumab levels in 15 samples within 123 days after administration. A shift in the pembrolizumab clearance rate was evident ensuing day 77 (0.6 µg/mL) after administration. Pembrolizumab levels up to day 77 (9.1-0.6 µg/mL) strongly exhibited a linear, first-order clearance (R2 = 0.991), whereas after day 77, an accelerated non-linear clearance was observed. This transition from a linear to non-linear clearance was most likely a result of full target receptor saturation to non-full target receptor saturation, in which the added effect of target-mediated drug disposition occurs. This suggests that pembrolizumab's targets were fully saturated at levels above 0.6 µg/mL, which is 43 to 61 times lower than the steady-state trough levels (Ctrough,ss) of the currently registered fixed-dosing regimens (3-5).
Collapse
Affiliation(s)
- Fenna de Vries
- Department of Pharmacy, OLVG Hospital, Amsterdam, Netherlands,*Correspondence: Fenna de Vries,
| | | | - Gertjan Wolbink
- Department of Rheumatology, Amsterdam Rheumatology and Immunology Centre, Amsterdam, Netherlands
| | - Annick de Vries
- Diagnostic Services, Sanquin Health Solutions, Amsterdam, Netherlands
| | - Floris C. Loeff
- Diagnostic Services, Sanquin Health Solutions, Amsterdam, Netherlands
| | | |
Collapse
|
26
|
Lu Y, Shimizu S, Sawamura R, Tajima N, He L, Lee M, Abutarif M, Shi R. Population Pharmacokinetics of Patritumab Deruxtecan in Patients With Solid Tumors. J Clin Pharmacol 2023; 63:77-90. [PMID: 36053771 PMCID: PMC10087001 DOI: 10.1002/jcph.2137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022]
Abstract
Patritumab deruxtecan is an antibody-drug conjugate consisting of a fully human monoclonal antibody against human epidermal growth factor receptor 3 (HER3) attached to a topoisomerase I inhibitor payload via a tetrapeptide-based cleavable linker. As part of the pharmacometric analysis informing dose selection for later-stage development, population pharmacokinetics (PK) analysis of patritumab deruxtecan was conducted with pooled serum PK data from patients with HER3-expressing solid tumors (from 3 phase 1/2 studies in breast, lung, and colorectal cancer; N = 425) treated over the dose range of 1.6 to 8.0 mg/kg intravenously every 3 weeks. Population PK modeling for deruxtecan (DXd)-conjugated antibody (representing patritumab deruxtecan) and unconjugated MAAA-1181a (DXd, payload) was carried out sequentially. DXd-conjugated antibody PK was described using a 2-compartment model with parallel linear and nonlinear clearance. Unconjugated DXd PK was described using a 1-compartment model with linear clearance and release of DXd as a first-order, time-dependent function of the level of DXd-conjugated antibody in the central compartment. Preliminary covariate evaluation was conducted for prespecified covariates of pharmacological plausibility and clinical interest. The final model retained weight (on linear clearance and central volume) and albumin level, sex, and tumor type (on linear clearance) for DXd-conjugated antibody, and weight (on release rate constant) and hepatic function (on clearance) for unconjugated DXd. Effects of these covariates on the exposure metrics were generally mild and did not require dose adjustment for subpopulations in subsequent development. Further PK characterization for patritumab deruxtecan will evolve with emerging data.
Collapse
Affiliation(s)
- Yasong Lu
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | | | | | | | - Ling He
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | - Mark Lee
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | | | - Rong Shi
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| |
Collapse
|
27
|
Zheng M. Serum albumin: a pharmacokinetic marker for optimizing treatment outcome of immune checkpoint blockade. J Immunother Cancer 2022; 10:jitc-2022-005670. [PMID: 36600664 PMCID: PMC9772729 DOI: 10.1136/jitc-2022-005670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2022] [Indexed: 12/24/2022] Open
Abstract
As we look forward to the bright future of immune checkpoint blockade (ICB) therapy, there is still lacking a pharmacokinetic marker to understand the inter-individual differences in ICB response. ICB therapy is based on IgG antibodies that share the same homeostatic pathway with serum albumin. Therefore, serum albumin level could reflect IgG catabolic rate that directly impacts the clearance of therapeutic IgG antibodies. Through interrogating a large, clinically representative pan-cancer cohort of 1,479 ICB-treated patients, this study found that higher baseline albumin levels were significantly associated with stepwise improvements in overall survival (OS), progression-free survival (PFS), and objective response rate (ORR) (p<0.001), with the variability and reproducibility confirmed in 1,000 bootstrap-resampled cohorts. Furthermore, these findings were also confirmed in most subgroups defined by patient demographics, baseline characteristics, treatments, and cancer types, even in those with low ICB-responsive cancer types and low tumor mutation burden (TMB) (TMB≤10 mut/Mb) that most of which have not been approved by the US Food and Drug Administration (FDA) for ICB therapy. In summary, this study highlights the importance of pretreatment pharmacokinetic modeling for predicting ICB treatment outcomes. Based on serum albumin-an inexpensive, non-invasive, and easily accessible biomarker of IgG pharmacokinetics, we could take a step further towards optimizing ICB therapy.
Collapse
Affiliation(s)
- Ming Zheng
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, 27 Taiping Road, Beijing 100850, China,Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| |
Collapse
|
28
|
Mazzaschi G, Quaini F, Buti S. Exploring genetic and immune underpinnings of the sexual dimorphism in tumor response to immune checkpoints inhibitors: A narrative review. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 4:100146. [PMID: 36571078 PMCID: PMC9772791 DOI: 10.1016/j.crphar.2022.100146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction In spite of the undisputed relevance of sex as critical biologic variable of the immune landscape, still limited is our understanding of the basic mechanisms implicated in sex-biased immune response thereby conditioning the therapeutic outcome in cancer patients. This hindrance delays the actual attempts to decipher the heterogeneity of cancer and its immune surveillance, further digressing the achievement of predictive biomarkers in the current immunotherapy-driven scenario. Body: The present review concisely reports on genetic, chromosomal, hormonal, and immune features underlying sex-differences in the response to immune checkpoint inhibitors (ICIs). In addition to outline the need of robust data on ICI pharmaco-kinetics/dynamics, our survey might provide new insights on sex determinants of ICI efficacy and suggests uncovered pathways that warrant prospective investigations. Conclusion According to a sharable view, we propose to widely include sex among the co-variates when assessing the clinical response to ICI in cancer patients.
Collapse
Affiliation(s)
- Giulia Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
29
|
Hayato S, Takenaka O, Sreerama Reddy SH, Landry I, Reyderman L, Koyama A, Swanson C, Yasuda S, Hussein Z. Population pharmacokinetic-pharmacodynamic analyses of amyloid positron emission tomography and plasma biomarkers for lecanemab in subjects with early Alzheimer's disease. CPT Pharmacometrics Syst Pharmacol 2022; 11:1578-1591. [PMID: 36165093 PMCID: PMC9755918 DOI: 10.1002/psp4.12862] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/08/2022] [Accepted: 08/03/2022] [Indexed: 11/06/2022] Open
Abstract
Lecanemab is a humanized immunoglobulin G1 monoclonal antibody that selectively binds to soluble Aβ aggregate species, while demonstrating low affinity for Aβ monomer. This article describes the population pharmacokinetic (PK) and PK/pharmacodynamic (PD) analyses for amyloid plaques, as measured using positron emission tomography (PET), and biomarkers of amyloid pathology as evidenced by Aβ42/40 ratio and plasma p-tau181 following i.v. administration of lecanemab in subjects with early Alzheimer's disease. Lecanemab PKs were well-characterized with a two-compartment model with first-order elimination. Final PK model contained covariate effects of anti-drug antibody positive status, sex, body weight, and albumin on clearance. The time course of amyloid PET standard uptake ratio (SUVr), plasma Aβ42/40 ratio, and p-tau181 were described using indirect response models with lecanemab exposure as a maximum effect function stimulating the reduction of SUVr, and as a linear function increasing Aβ42/40 ratio and decreasing p-tau181 formation rates. PK/PD simulations show that 10 mg/kg biweekly dosing results in larger and faster decrease in SUVr and p-tau181 and increase in Aβ42/40 ratio as compared to 10 mg/kg monthly dose. Furthermore, the PK/PD simulations showed that after treatment discontinuation the brain amyloid re-accumulation to baseline levels is slow with a recovery half-life of ~4 years, whereas plasma Aβ42/40 ratio and p-tau181 return to baseline levels faster than amyloid. Given the relationship between changes in amyloid PET SUVr and soluble biomarkers, the developed PK/PD models can be used to inform lecanemab dose regimens in future clinical studies.
Collapse
|
30
|
Likic R, Dear JW, Lewis LD. Spotlight commentary: Navigating between financial risks and improved treatment outcomes with immune checkpoint inhibitors in cancer patients: The need for biomarker identification and dose optimization. Br J Clin Pharmacol 2022; 88:5060-5063. [PMID: 36206769 DOI: 10.1111/bcp.15548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Robert Likic
- University of Zagreb Medical School, Zagreb, Croatia.,Department of Internal Medicine, Division of Clinical Pharmacology and Therapeutics, Clinical Hospital Centre Zagreb, Zagreb, Croatia
| | - James W Dear
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Lionel D Lewis
- Section of Clinical Pharmacology, Dept. of Medicine, The Geisel School of Medicine & Dartmouth-Hitchcock Medical Centre, Lebanon, New Hampshire, USA
| |
Collapse
|
31
|
Shang J, Huang L, Huang J, Ren X, Liu Y, Feng Y. Population pharmacokinetic models of anti-PD-1 mAbs in patients with multiple tumor types: A systematic review. Front Immunol 2022; 13:871372. [PMID: 35983041 PMCID: PMC9379304 DOI: 10.3389/fimmu.2022.871372] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/28/2022] [Indexed: 11/20/2022] Open
Abstract
Aims and background A number of population pharmacokinetic (PPK) models of anti-programmed cell death-1 (PD-1) monoclonal antibodies (mAbs) in multiple tumor types have been published to characterize the influencing factors of their pharmacokinetics. This review described PPK models of anti-PD-1 mAbs that investigate the magnitude and types of covariate effects in PK parameters, provide a reference for building PPK models of other anti-PD-1 mAbs, and identify areas requiring additional research to facilitate the application of PPK models. Methods A systematic search for analyses of PPK models of eleven anti-PD-1 mAbs on the market that were carried out in humans was conducted using PubMed, Embase, and the Cochrane Library. The search covered the period from the inception of the databases to April 2022. Results Currently, there are fourteen analyses on PPK models of anti-PD-1 mAbs summarized in this review, including seven models that refer to nivolumab, four referring to pembrolizumab, one referring to cemiplimab, one referring to camrelizumab, and one referred to dostarlimab. Most analyses described the pharmacokinetics of anti-PD-1 mAbs with a two-compartment model with time-varying clearance (CL) and a sigmoidal maximum effect. The estimated CL and volume of distribution in the central (VC) ranged from 0.179 to 0.290 L/day and 2.98 to 4.46 L, respectively. The median (range) of interindividual variability (IIV) for CL and VC was 30.9% (8.7%–50.8%) and 29.0% (4.32%–40.7%), respectively. The commonly identified significant covariates were body weight (BW) on CL and VC, and albumin (ALB), tumor type, sex, and performance status (PS) on CL. Other less assessed significant covariates included lactate dehydrogenase (LDH), immunoglobulin G (IgG), ipilimumab coadministration (IPICO) on CL, and body mass index (BMI), malignant pleural mesothelioma (MESO) on VC. Conclusion This review provides detailed information about the characteristics of PPK models of anti-PD-1 mAbs, the effects of covariates on PK parameters, and the current status of the application of the models. ALB, BW, specific tumor type, sex, and PS should be considered for the future development of the PPK model of anti-PD-1 mAbs. Other potential covariates that were assessed less frequently but still have significance (e.g., LDH, IgG, and IPICO) should not be ignored. Thus, further research and thorough investigation are needed to assess new or potential covariates, which will pave the way for personalized anti-PD-1 mAbs therapy.
Collapse
Affiliation(s)
- Jingyuan Shang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China.,Faculty of Life Sciences and Biopharmaceuticals, Shenyang Pharmceutical University, Shenyang, China
| | - Lin Huang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Jing Huang
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Xiaolei Ren
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Yi Liu
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Yufei Feng
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| |
Collapse
|
32
|
Soehoel A, Larsen MS, Timmermann S. Population Pharmacokinetics of Tralokinumab in Adult Subjects With Moderate to Severe Atopic Dermatitis. Clin Pharmacol Drug Dev 2022; 11:910-921. [PMID: 35671038 PMCID: PMC9796478 DOI: 10.1002/cpdd.1113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/02/2022] [Indexed: 01/01/2023]
Abstract
Tralokinumab is the first biologic therapy for moderate-to-severe atopic dermatitis (AD) that specifically neutralizes interleukin-13 activity, a key driver of AD signs and symptoms. Tralokinumab is a human immunoglobulin G4 monoclonal antibody administered subcutaneously every 2 weeks (with possibility of maintenance dosing every 4 weeks). This population pharmacokinetic (PK) analysis aimed to identify sources of PK variability and relevant predictors of tralokinumab exposure in adults with moderate to severe AD. Nonlinear mixed-effect modeling, including covariate analysis, was used on a data set including 2561 subjects (AD, asthma, healthy) from 10 clinical trials. A 2-compartment model with first-order absorption and elimination adequately described the tralokinumab PK. Body weight was identified as a relevant predictor of tralokinumab exposure; other covariates including age, sex, race, ethnicity, disease type, AD severity, and renal and hepatic impairment were not. For body weight, the difference in exposure between the upper- and lower-weight quartiles in patients with AD was <2-fold, supporting the appropriateness of flat dosing (300 mg). Given the reduced exposure associated with higher body weight, coupled with the reduced exposure provided by dosing every 4 weeks, it is uncertain whether higher-weight patients will achieve sufficient exposure to maintain efficacy if dosed every 4 weeks instead of the standard every 2 weeks.
Collapse
Affiliation(s)
| | - Malte Selch Larsen
- Clinical PharmacologyLEO Pharma A/SBallerupDenmark,Present address:
Novo NordiskSøborgDenmark
| | | |
Collapse
|
33
|
Guo Y, Wei L, Patel SH, Lopez G, Grogan M, Li M, Haddad T, Johns A, Ganesan LP, Yang Y, Spakowicz DJ, Shields PG, He K, Bertino EM, Otterson GA, Carbone DP, Presley C, Kulp SK, Mace TA, Coss CC, Phelps MA, Owen DH. Serum Albumin: Early Prognostic Marker of Benefit for Immune Checkpoint Inhibitor Monotherapy But Not Chemoimmunotherapy. Clin Lung Cancer 2022; 23:345-355. [PMID: 35131184 PMCID: PMC9149057 DOI: 10.1016/j.cllc.2021.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/07/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cancer cachexia exhibits decreased albumin and associates with short overall survival (OS) in patients with non-small cell lung cancer (NSCLC), but whether on-treatment albumin changes associate with OS in NSCLC patients treated with immune checkpoint inhibitors (ICIs) and combination chemoimmunotherapy has not been thoroughly evaluated. PATIENTS AND METHODS We conducted a single-center retrospective study of patients with advanced NSCLC who received first-line ICI with or without chemotherapy between 2013 and 2020. The association of pretreatment albumin and early albumin changes with OS was evaluated using Kaplan-Meier method and Cox regression models. RESULTS A total of 210 patients were included: 109 in ICI cohort and 101 in ICI + Chemo cohort. Within a median of 21 days from treatment initiation, patients with ≥ 10% of albumin decrease had significantly shorter OS compared to patients without albumin decrease in ICI cohort. Pretreatment albumin and albumin decrease within the first or second cycle of treatment were significantly and independently associated with OS in ICI cohort, but not in ICI + Chemo cohort. The lack of association between albumin and OS with the addition of chemotherapy was more pronounced among patients with ≥ 1% PD-L1 expression in subgroup analysis. CONCLUSION Pretreatment serum albumin and early albumin decrease in ICI monotherapy was significantly associated with OS in advanced NSCLC. Early albumin change, as a routine lab value tested in clinic, may be combined with established biomarkers to improve outcome predictions of ICI monotherapy. The underlying mechanism of the observed association between decreased albumin and ICI resistance warrants further investigation.
Collapse
Affiliation(s)
- Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Lai Wei
- Center for Biostatistics, Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH
| | - Sandip H Patel
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Gabrielle Lopez
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Madison Grogan
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Mingjia Li
- Department of Internal Medicine, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Tyler Haddad
- Department of Internal Medicine, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Andrew Johns
- Department of Internal Medicine, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Latha P Ganesan
- Department of Internal Medicine, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Yiping Yang
- Division of Hematology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Daniel J Spakowicz
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Peter G Shields
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Kai He
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Erin M Bertino
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Gregory A Otterson
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - David P Carbone
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Carolyn Presley
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Samuel K Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A Mace
- 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.
| | - Dwight H Owen
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH.
| |
Collapse
|
34
|
Maritaz C, Broutin S, Chaput N, Marabelle A, Paci A. Immune checkpoint-targeted antibodies: a room for dose and schedule optimization? J Hematol Oncol 2022; 15:6. [PMID: 35033167 PMCID: PMC8760805 DOI: 10.1186/s13045-021-01182-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-CTLA-4 and anti-PD-1/PD-L1 immune checkpoint inhibitors are therapeutic monoclonal antibodies that do not target cancer cells but are designed to reactivate or promote antitumor immunity. Dosing and scheduling of these biologics were established according to conventional drug development models, even though the determination of a maximum tolerated dose in the clinic could only be defined for anti-CTLA-4. Given the pharmacology of these monoclonal antibodies, their high interpatient pharmacokinetic variability, the actual clinical benefit as monotherapy that is observed only in a specific subset of patients, and the substantial cost of these treatments, a number of questions arise regarding the selected dose and the dosing interval. This review aims to outline the development of these immunotherapies and considers optimization options that could be used in clinical practice.
Collapse
Affiliation(s)
- Christophe Maritaz
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sophie Broutin
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Chaput
- Laboratory for Immunomonitoring in Oncology (LIO), Faculty of Pharmacy, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Aurélien Marabelle
- Drug Development Unit (DITEP), LRTI U1015 INSERM, Gustave Roussy, Villejuif, France
| | - Angelo Paci
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France.
- Pharmacokinetic Unit, Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.
| |
Collapse
|
35
|
Xiang H, Liu L, Gao Y, Ahene A, Collins H. Covariate effects and population pharmacokinetic analysis of the anti-FGFR2b antibody bemarituzumab in patients from phase 1 to phase 2 trials. Cancer Chemother Pharmacol 2021; 88:899-910. [PMID: 34383128 PMCID: PMC8484135 DOI: 10.1007/s00280-021-04333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/17/2021] [Indexed: 11/25/2022]
Abstract
Purpose A population pharmacokinetic (PK) analysis of the anti-fibroblast growth factor receptor 2b antibody, bemarituzumab, was performed to evaluate the impact of covariates on the PK and assess whether dose adjustment is necessary for a future phase 3 trial. Methods Serum concentration data were obtained from three clinical trials, with 1552 bemarituzumab serum samples from 173 patients, and were analyzed using nonlinear mixed-effects modeling. Results A two-compartment model with parallel linear and nonlinear (Michaelis–Menten) elimination from the central compartment best described the bemarituzumab serum concentration data. The final model estimated a typical linear clearance (CL) of 0.311 L/day, volume of distribution in the central compartment (Vc) of 3.58 L, distribution clearance (Q) of 0.952 L/day, volume of distribution in the peripheral compartment (Vp) of 2.71 L, maximum drug elimination by nonlinear clearance (Vmax) of 2.80 μg/day, and Michaelis–Menten constant (Km) of 4.45 μg/mL. Baseline body weight, baseline albumin, gender, and chemotherapy were identified as statistically significant covariates on the PK of bemarituzumab. Given the low interindividual variability of bemarituzumab key PK parameters (CL and Vc) and the small or modest effect of all statistically significant covariates on bemarituzumab exposure at steady-state, no covariate is expected to have clinically meaningful effects on bemarituzumab exposure. Conclusion No covariate had a clinically meaningful impact on bemarituzumab exposure. These results indicate that dose adjustment of bemarituzumab is not necessary, based on the aforementioned covariates, for a future phase 3 trial in gastric and gastroesophageal junction adenocarcinoma population with FGFR2b overexpression in combination with mFOLFOX6. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-021-04333-y.
Collapse
Affiliation(s)
- Hong Xiang
- Five Prime Therapeutics, Inc., South San Francisco, CA, USA. .,Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA.
| | - Lucy Liu
- Shanghai Qiangshi Information Technology Co., Ltd., Shanghai, China
| | - Yuying Gao
- Shanghai Qiangshi Information Technology Co., Ltd., Shanghai, China
| | - Ago Ahene
- Five Prime Therapeutics, Inc., South San Francisco, CA, USA.,Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Helen Collins
- Five Prime Therapeutics, Inc., South San Francisco, CA, USA.,Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| |
Collapse
|
36
|
Grinshpun B, Thorsteinson N, Pereira JN, Rippmann F, Nannemann D, Sood VD, Fomekong Nanfack Y. Identifying biophysical assays and in silico properties that enrich for slow clearance in clinical-stage therapeutic antibodies. MAbs 2021; 13:1932230. [PMID: 34116620 PMCID: PMC8204999 DOI: 10.1080/19420862.2021.1932230] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Understanding the pharmacokinetic (PK) properties of a drug, such as clearance, is a crucial step for evaluating efficacy. The PK of therapeutic antibodies can be complex and is influenced by interactions with the target, Fc-receptors, anti-drug antibodies, and antibody intrinsic factors. A growing body of literature has linked biophysical properties of antibodies, particularly nonspecific-binding propensity, hydrophobicity and charged regions to rapid clearance in preclinical species and selected human PK studies. A clear understanding of the connection between biophysical properties and their impact on PK would allow for early selection and optimization of antibodies and reduce costly attrition during clinical trials due to sub-optimal human clearance. Due to the difficulty in obtaining large and unbiased human PK data, previous studies have focused mostly on preclinical PK. For this study, we obtained and curated the most comprehensive clinical PK dataset to date and calculated accurate estimates of linear clearance for 64 monoclonal antibodies ranging from investigational candidates in Phase 2 trials to marketed products. This allows for the first time a deep analysis of the influence of biophysical and sequence-based in silico properties directly on human clearance. We use statistical analysis and a Random Forest classifier to identify properties that have the greatest influence in our dataset. Our findings indicate that in vitro poly-specificity assay and in silico estimated isoelectric point can discriminate fast and slow clearing antibodies, extending previous observations on preclinical clearance. This provides a simple yet powerful approach to select antibodies with desirable PK during early-stage screening.
Collapse
Affiliation(s)
- Boris Grinshpun
- Discovery & Development Technologies, Drug Disposition & Design, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| | - Nels Thorsteinson
- Department of Scientific Services, Chemical Computing Group ULC, Montreal, Quebec, Canada
| | - Joao Ns Pereira
- Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany
| | - Friedrich Rippmann
- Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany
| | - David Nannemann
- Discovery & Development Technologies, Drug Disposition & Design, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| | - Vanita D Sood
- Discovery & Development Technologies, Drug Disposition & Design, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| | - Yves Fomekong Nanfack
- Discovery & Development Technologies, Drug Disposition & Design, EMD Serono Research & Development Institute, Inc, Billerica, MA, USA
| |
Collapse
|
37
|
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
|
38
|
Passot C, Sberro-Soussan R, Bertrand D, Caillard S, Schvartz B, Domenger C, Contin-Bordes C, Paintaud G, Halimi JM, Ternant D, Gatault P. Feasibility and safety of tailored dosing schedule for eculizumab based on therapeutic drug monitoring: Lessons from a prospective multicentric study. Br J Clin Pharmacol 2020; 87:2236-2246. [PMID: 33118186 DOI: 10.1111/bcp.14627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 12/23/2022] Open
Abstract
AIMS Eculizumab is an anti-C5 monoclonal antibody approved for rare diseases including atypical haemolytic-uraemic syndrome. The maintenance phase dosing regimen is identical for all adult patients: 1200 mg every 2 weeks. Recent studies reported an overexposure in many patients when considering a target trough concentration range of 50-100 mg/L. The aim of the present work was to validate the feasibility of therapeutic drug monitoring of eculizumab in atypical haemolytic-uraemic syndrome patients. METHODS We performed a 2-step prospective multicentre study. In the first phase, we developed a pharmacokinetic population model using data from 40 patients and identified patients for whom a 1-week lengthening of interval between infusions would lead to a trough concentration above 100 mg/L. In the second phase, selected patients were allocated a 1-week extension and eculizumab trough concentrations were monitored. RESULTS The model confirmed the previously reported influence of bodyweight on elimination clearance and predicted that 36 (90%) patients would be eligible for interval extension. In the second phase of the study, a 1-week lengthening of interval between infusions was performed in 15 patients whose trough concentration at the next visit was predicted with a Bayesian model to be above 100 mg/L. After interval extension, 10 patients (67%) presented measured trough concentrations over 100 mg/L. No biological or clinical recurrence of disease was observed, even in the 5 patients with concentrations below 100 mg/L in whom the initial dosing regimen was resumed. CONCLUSION Safe eculizumab interval adjustment is feasible with a PK monitoring.
Collapse
Affiliation(s)
- Christophe Passot
- EA7501, University of Tours, France.,Laboratory of Pharmacology-Toxicology, CHRU de Tours, France.,Integrated Center for Oncology, Angers, France
| | - Rebecca Sberro-Soussan
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Sophie Caillard
- Nephrology and Transplant Department, Strasbourg University Hospital, Strasbourg, France
| | | | | | | | - Gilles Paintaud
- EA7501, University of Tours, France.,Laboratory of Pharmacology-Toxicology, CHRU de Tours, France
| | - Jean-Michel Halimi
- EA4245 Transplant Immunology and Inflammation, Université de Tours, Tours, France.,Service de Néphrologie-Hypertension artérielle, Dialyses, Transplantation rénale, CHRU Tours, France
| | - David Ternant
- EA7501, University of Tours, France.,Laboratory of Pharmacology-Toxicology, CHRU de Tours, France
| | - Philippe Gatault
- EA4245 Transplant Immunology and Inflammation, Université de Tours, Tours, France.,Service de Néphrologie-Hypertension artérielle, Dialyses, Transplantation rénale, CHRU Tours, France
| |
Collapse
|
39
|
Yin O, Xiong Y, Endo S, Yoshihara K, Garimella T, AbuTarif M, Wada R, LaCreta F. Population Pharmacokinetics of Trastuzumab Deruxtecan in Patients With HER2-Positive Breast Cancer and Other Solid Tumors. Clin Pharmacol Ther 2020; 109:1314-1325. [PMID: 33118153 PMCID: PMC8246728 DOI: 10.1002/cpt.2096] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/17/2020] [Indexed: 12/26/2022]
Abstract
Trastuzumab deruxtecan (DS‐8201) is a human epidermal growth factor receptor 2 (HER2)–targeting antibody–drug conjugate with a novel enzyme‐cleavable linker, a topoisomerase I inhibitor payload, and a drug‐to‐antibody ratio of ≈ 8. We have characterized the population pharmacokinetics (PK) of trastuzumab deruxtecan and released drug (topoisomerase I inhibitor) in patients with HER2‐positive breast cancer or other solid tumor malignancies. This analysis includes pooled data from five clinical studies with 639 patients. Trastuzumab deruxtecan doses ranged from 0.8 to 8.0 mg/kg every 3 weeks. Serum concentrations of trastuzumab deruxtecan and released drug were analyzed using a sequential two‐step approach, with the nonlinear mixed‐effects modeling methods. Covariate assessment was based upon stepwise forward‐addition and backward‐elimination process, followed by both univariate and multivariate analysis quantifying their impact on steady‐state exposure of trastuzumab deruxtecan and released drug. A two‐compartment model with linear elimination best described PK profiles of intact trastuzumab deruxtecan, while a one‐compartment model with time‐varying release‐rate constant and linear elimination described released‐drug PK profiles. Statistically significant covariates (country, tumor size, sex, formulation, age, body weight, albumin, total bilirubin, and aspartate aminotransferase) resulted in < 20% change in steady‐state area under the concentration‐time curve of trastuzumab deruxtecan and released drug, except for increased body weight (95th percentile, 86 kg) and decreased albumin (5th percentile, 31 g/L). Analysis of patients stratified by country, race, renal function, and hepatic function found no clinically meaningful differences in steady‐state exposure of intact trastuzumab deruxtecan or released drug. Overall, results suggest that no dose adjustment based on tested covariates or in specific patient populations is warranted.
Collapse
Affiliation(s)
- Ophelia Yin
- Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc, Basking Ridge, New Jersey, USA
| | - Yuan Xiong
- Certara Strategic Consulting, Princeton, New Jersey, USA
| | - Seiko Endo
- Clinical Pharmacology Department, Daiichi Sankyo Co, Ltd, Tokyo, Japan
| | | | - Tushar Garimella
- Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc, Basking Ridge, New Jersey, USA
| | - Malaz AbuTarif
- Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc, Basking Ridge, New Jersey, USA
| | - Russ Wada
- Certara Strategic Consulting, Princeton, New Jersey, USA
| | - Frank LaCreta
- Quantitative Clinical Pharmacology and Translational Sciences, Daiichi Sankyo, Inc, Basking Ridge, New Jersey, USA
| |
Collapse
|
40
|
Wang X, Du W, Zhang X, Li P. The Influence of Different Disease States on Rituximab Pharmacokinetics. Curr Drug Metab 2020; 21:938-946. [PMID: 32682367 DOI: 10.2174/1389200221666200719004035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/04/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The anti-CD20 antibody rituximab, which promotes the selective depletion of CD20 positive B cells, was the first targeted therapy that was approved for the treatment of B-cell malignancies, and it is now widely prescribed in both malignant and non-malignant, immune-related diseases. However, the cause of its various clinical responses in certain diseases, have not been clearly elucidated. The variabilities in inter-individual pharmacokinetic and the emerging evidence of the relationships between pharmacokinetic and pharmacodynamic may provide a better understanding of this drug. METHODS We searched and summarized the latest published articles on rituximab pharmacokinetic profiles and the pharmacokinetic/pharmacodynamic models in different patient populations, including B-cell malignancies, rheumatoid arthritis, ANCA-associated vasculitis, and glomerular kidney diseases. RESULTS Most pharmacokinetic data are drawn from clinical studies in oncology clinical practice. Body weight, gender, and antigen-related factors are proven to be the key factors affecting rituximab pharmacokinetics. In addition, the positive exposure-response relations were reported, which provide encouraging evidence for individualized therapies. While in immune disorders, especially in the off-labeled indications, pharmacokinetic studies are quite limited. Compared with that in B-cell malignancies, the differences in the pharmacokinetic parameters may be attributed to the different pathogeneses of diseases, mechanisms of action and dosing strategies. However, the correlation between drug exposure and clinical outcomes remains unclear. CONCLUSION Here, we provide an overview of the complexities associated with rituximab pharmacokinetics and pharmacodynamics in different diseases. Although many influencing factors need to be verified in future studies, a better understanding of the relationships between pharmacokinetic and pharmacodynamic may assist in optimizing rituximab clinical practice.
Collapse
Affiliation(s)
- Xiaoxing Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Wenwen Du
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xianglin Zhang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Pengmei Li
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
41
|
Bajaj G, Suryawanshi S, Roy A, Gupta M. Evaluation of covariate effects on pharmacokinetics of monoclonal antibodies in oncology. Br J Clin Pharmacol 2019; 85:2045-2058. [PMID: 31140642 DOI: 10.1111/bcp.13996] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 03/25/2019] [Accepted: 05/07/2019] [Indexed: 12/13/2022] Open
Abstract
AIMS The development of monoclonal antibodies (mAbs) requires an understanding of the interindividual variability (IIV) in pharmacokinetics (PK) at the population level facilitated by population PK (PopPK) modelling. However, there is no clear rationale for selecting which covariates to screen during PopPK model development. Here, we compare the effect of covariates on PK parameters for mAbs in oncology and identify the most commonly used covariates affecting PK parameters. METHODS All 25 mAbs approved for therapeutic use in oncology until December 2017 by the Food and Drug Administration and the European Medicines Agency were selected for study. Literature searches revealed 23 available PopPK models for these mAbs. To understand the magnitude and types of covariate effect on PK parameters, all covariates included in the final PopPK model for each mAb were summarized. RESULTS The most commonly identified covariates were baseline body weight (BW; 17 mAbs), baseline serum albumin (8 mAbs), and sex (7 mAbs) on clearance; and BW (16 mAbs) and sex (12 mAbs) on central volume of distribution. A reduced PopPK model was developed for nivolumab and ipilimumab using these covariates, and the percentage of explained IIV from the reduced model (20.3% and 16.8%, respectively) was compared with that from the full model (24.5% and 27.9%, respectively). CONCLUSIONS This analysis provides a uniform platform for selecting covariates and suggests that the effect of BW, albumin and sex should be included during the development of PopPK models for mAbs in oncology. The reduced model was able to explain IIV to a similar extent as the full model.
Collapse
Affiliation(s)
| | | | - Amit Roy
- Bristol-Myers Squibb, Princeton, NJ, USA
| | | |
Collapse
|