1
|
Delahousse J, Wagner AD, Borchmann S, Adjei AA, Haanen J, Burgers F, Letsch A, Quaas A, Oertelt-Prigione S, Özdemir BC, Verhoeven RHA, Della Pasqua O, Paci A, Mir O. Sex differences in the pharmacokinetics of anticancer drugs: a systematic review. ESMO Open 2024; 9:104002. [PMID: 39662226 PMCID: PMC11697095 DOI: 10.1016/j.esmoop.2024.104002] [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: 06/11/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND In addition to the effect of body weight, a patient's sex can influence the pharmacokinetics (PK) of anticancer agents, and thereby their activity and safety. The magnitude and relevance of sex differences, however, are currently unclear. METHODS We carried out a systematic review of published studies (clinical, n ≥ 10) on Food and Drug Administration (FDA)-approved (on 31 January 2022) anticancer drugs (excluding hormonal agents), aiming to identify significant PK differences between male and female patients. A difference of ≥20% on PK parameters (clearance or trough concentration) was considered significant. The methodological quality was assessed using the National Institutes of Health study quality assessment tool. This systematic review was conducted according to the PRISMA2020 guidelines and a previously published protocol, which was registered in the PROSPERO database (number 291008). RESULTS Data on 99 anticancer agents (for a total of 1643 abstracts and European Medicines Agency/FDA documents) were screened. The final dataset included 112 articles and 8 European Medicines Agency/FDA documents. The median size of a study cohort was 445 patients (range: 12-6468 patients). Significant PK differences (>+20% in clearance or apparent clearance in women) were identified for 14 drugs, and potentially significant PK differences (due to conflicting reports) for another 8 drugs. None of the studies included sex-based summaries to assess whether the observed differences in PK may impact the efficacy or safety profile. CONCLUSIONS Significant sex differences in PK have been identified including commonly used drugs of different classes, such as 5-fluorouracil, doxorubicin, paclitaxel, regorafenib, atezolizumab, and temozolomide. The risk-benefit ratio for such anticancer drugs is likely to be improved by the development of sex-specific dosing strategies. Additional sex-based PK-pharmacodynamic analyses are recommended during dose optimisation and are to be conducted in line with the FDA Project Optimus guidance. They should be reported even if no association between the patients' sex and the activity and/or toxicity of an anticancer drug has been identified.
Collapse
Affiliation(s)
- J Delahousse
- Department of Pharmacology, Gustave Roussy, Villejuif, France
| | - A D Wagner
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - S Borchmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Medical Faculty and University Hospital Cologne, Cologne, Germany; Cancer Center Cologne Essen (CCCE), Cologne, Germany; German Hodgkin Study Group, Cologne, Germany
| | - A A Adjei
- Taussig Cancer Center, Cleveland Clinic, Cleveland, USA
| | - J Haanen
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands; Melanoma Clinic, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - F Burgers
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - A Letsch
- Department of Hematology and Oncology, University Hospital Schleswig Holstein/University Cancer Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - A Quaas
- Institute of Pathology, University Hospital Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - S Oertelt-Prigione
- Gender Unit, Department of Primary and Community Care, Radboud University Medical Center, Nijmegen, Netherlands; AG10 Sex- and Gender-Sensitive Medicine, Medical Faculty OWL, University of Bielefeld, Bielefeld, Germany
| | - B C Özdemir
- Department of Medical Oncology, Bern University Hospital, Bern, Switzerland
| | - R H A Verhoeven
- Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, Netherlands; Department of Medical Oncology, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands; Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, Netherlands
| | - O Della Pasqua
- Clinical Pharmacology & Therapeutics Group, University College London, London, UK
| | - A Paci
- Department of Pharmacology, Gustave Roussy, Villejuif, France; Pharmacokinetics Department, Faculté de Pharmacie, Université Paris-Saclay, Gif-sur-Yvette, France
| | - O Mir
- Department of Pharmacology, Gustave Roussy, Villejuif, France
| |
Collapse
|
2
|
Wang K, Pan C, Xu F, Tse AN, Sheng Y. Comprehensive population pharmacokinetic modelling of sugemalimab, an anti-programmed death-ligand 1 (PD-L1) human monoclonal antibody, in patients with solid tumours or lymphomas across multiple Phase I-III studies. Br J Clin Pharmacol 2024. [PMID: 39389094 DOI: 10.1111/bcp.16276] [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: 03/08/2024] [Revised: 09/02/2024] [Accepted: 09/15/2024] [Indexed: 10/12/2024] Open
Abstract
AIMS The aim of this study was to develop a population pharmacokinetics model for sugemalimab, a monoclonal antibody that targets programmed death-ligand 1 (PD-L1), using data from Phase I-III trials and to assess clinical factors affecting sugemalimab exposure. METHODS A nonlinear mixed-effect modelling approach was employed to analyse pooled data from nine studies involving 1628 subjects to characterize the PopPK of sugemalimab. This investigation examined the influence of various covariates on sugemalimab pharmacokinetics (PK), encompassing demographics, baseline hepatic and renal function-related covariates, and others (including anti-drug antibody [ADA], combination treatment, Eastern Cooperative Oncology Group [ECOG] performance score, tumour burden and tumour type). Estimation accuracy and predictive ability of the final model were evaluated using various methods. The influence of covariates on sugemalimab exposure was assessed by simulation from the final model. RESULTS A two-compartment model with first-order elimination and time-varying clearance effectively described the PK of sugemalimab. Covariate analyses revealed significant relationships between sugemalimab clearance and body weight, albumin, gender, ADA, tumour burden and tumour type. The statistically significant covariates on central volume were body weight, albumin, gender and tumour type. No significant relationships were found in the final model for age, race, alanine aminotransferase, aspartate aminotransferase, creatinine, total bilirubin, alkaline phosphatase, combination treatment, creatinine clearance, ECOG, renal function or hepatic function. All significant covariates demonstrated less than a 20% effect on sugemalimab exposure. CONCLUSIONS The PopPK model adequately described the pharmacokinetic profile of sugemalimab with no clinically meaningful impact observed on its exposure across all covariates. Dose adjustment does not appear to be necessary.
Collapse
Affiliation(s)
- Kun Wang
- Shanghai Qiangshi Information Technology Co., Ltd., Shanghai, China
| | - Chaohsuan Pan
- Cstone Pharmaceuticals (Suzhou) Co., Ltd., Shanghai, China
| | - Fengyan Xu
- Shanghai Qiangshi Information Technology Co., Ltd., Shanghai, China
| | - Archie N Tse
- Cstone Pharmaceuticals (Suzhou) Co., Ltd., Shanghai, China
| | - Yucheng Sheng
- Cstone Pharmaceuticals (Suzhou) Co., Ltd., Shanghai, China
| |
Collapse
|
3
|
Shemesh CS, Wang Y, An A, Ding H, Chan P, Liu Q, Chen YW, Wu B, Wu Q, Wang X. Phase I pharmacokinetic, safety, and preliminary efficacy study of tiragolumab in combination with atezolizumab in Chinese patients with advanced solid tumors. Cancer Chemother Pharmacol 2024; 94:45-55. [PMID: 38451273 PMCID: PMC11258083 DOI: 10.1007/s00280-024-04650-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: 12/21/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Tiragolumab is an immunoglobulin G1 monoclonal antibody targeting the immune checkpoint T cell immunoreceptor with immunoglobulin and immunoreceptor ITIM domains. Targeting multiple immune pathways may improve anti-tumor responses. The phase I YP42514 study assessed the pharmacokinetics (PK), safety, and preliminary efficacy of tiragolumab plus atezolizumab in Chinese patients with advanced solid tumors. METHODS Adult patients from mainland China with Eastern Cooperative Oncology Group performance score 0/1, life expectancy of ≥ 12 weeks, and adequate hematologic/end organ function were eligible. Patients received tiragolumab 600 mg and atezolizumab 1200 mg intravenous every 3 weeks. Key endpoints were PK (serum concentrations of tiragolumab and atezolizumab) and safety. Results from this study were compared with the global phase I study, GO30103 (NCT02794571). RESULTS In this study, 20 patients received a median of five doses of tiragolumab plus atezolizumab. Median age was 57.5 years, 85.0% of patients were male and the most common tumor type was non-small cell lung cancer. Exposures in Chinese patients were comparable to the global GO30103 population: geometric mean ratio was 1.07 for Cycle 1 tiragolumab area under the concentration-time curve0-21 and 0.92 and 0.93 for Cycle 1 peak and trough atezolizumab exposure, respectively. Treatment-related adverse events were consistent across the Chinese and global populations. Two patients (10.0%) in this study achieved a partial response. CONCLUSION In this study, tiragolumab plus atezolizumab was tolerable and demonstrated preliminary anti-tumor activity. There were no meaningful differences in the PK or safety of tiragolumab plus atezolizumab between the Chinese and global populations. CLINICAL TRIAL REGISTRATION NUMBER China Clinical Trial Registry Identifier CTR20210219/YP42514. Date of registration 16 March 2021.
Collapse
Affiliation(s)
- Colby S Shemesh
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA.
| | - Yongsheng Wang
- Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, China
| | - Andrew An
- Safety Science, F. Hoffmann-La Roche Ltd, Beijing, China
| | - Hao Ding
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Phyllis Chan
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Qi Liu
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Yih-Wen Chen
- Bioanalytical Science, Genentech Inc., South San Francisco, CA, USA
| | - Benjamin Wu
- Clinical Pharmacology, Genentech Inc., South San Francisco, CA, USA
| | - Qiong Wu
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Shanghai, China
| | - Xian Wang
- Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
Puszkiel A, Bianconi G, Pasquiers B, Balakirouchenane D, Arrondeau J, Boudou-Rouquette P, Bretagne MC, Salem JE, Declèves X, Vidal M, Kramkimel N, Guegan S, Aractingi S, Huillard O, Alexandre J, Wislez M, Goldwasser F, Blanchet B. Extending the dosing intervals of nivolumab: model-based simulations in unselected cancer patients. Br J Cancer 2024; 130:1866-1874. [PMID: 38532102 PMCID: PMC11130267 DOI: 10.1038/s41416-024-02659-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Reducing nivolumab dose intensity could increase patients' life quality and decrease the financial burden while maintaining efficacy. The aims of this study were to develop a population PK model of nivolumab based on data from unselected metastatic cancer patients and to simulate extended-interval regimens allowing to maintain minimal effective plasma concentrations (MEPC). METHODS Concentration-time data (992 plasma nivolumab concentrations, 364 patients) were modeled using a two-compartment model with linear elimination clearance in Monolix software. Extended-interval regimens allowing to maintain steady-state trough concentrations (Cmin,ss) above the MEPC of 2.5 mg/L or 1.5 mg/L in >90% of patients were simulated. RESULTS Increasing 3-times the dosing interval from 240 mg every two weeks (Q2W) to Q6W and 2-times from 480 mg Q4W to Q8W resulted in Cmin,ss above 2.5 mg/L in 95.8% and 95.4% of patients, respectively. 240 mg Q8W and 480 mg Q10W resulted in Cmin,ss above 1.5 mg/L in 91.0% and 91.8% of patients, respectively. Selection of a 240 mg Q6W regimen would decrease by 3-fold the annual treatment costs compared to standard regimen of 240 mg Q2W (from 78,744€ to 26,248€ in France). CONCLUSIONS Clinical trials are warranted to confirm the non-inferiority of extended-interval compared to standard regimen.
Collapse
Affiliation(s)
- Alicja Puszkiel
- Université Paris Cité, Faculté de Pharmacie de Paris, INSERM UMR-S1144, Paris, France.
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France.
| | - Guillaume Bianconi
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
| | - Blaise Pasquiers
- Université Paris Cité, Faculté de Pharmacie de Paris, INSERM UMR-S1144, Paris, France
- PhinC Development, Massy, France
| | | | - Jennifer Arrondeau
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
| | - Pascaline Boudou-Rouquette
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
| | - Marie-Claire Bretagne
- Department of Pharmacology, Pharmacovigilance Unit, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Joe-Elie Salem
- Department of Pharmacology, Pharmacovigilance Unit, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
- INSERM, CIC-1901, Sorbonne Université, Paris, France
| | - Xavier Declèves
- Université Paris Cité, Faculté de Pharmacie de Paris, INSERM UMR-S1144, Paris, France
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
| | - Michel Vidal
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
- Université Paris Cité, Faculté de Pharmacie de Paris, UMR8038 CNRS CiTCoM, U1268 INSERM, CARPEM, Paris, France
| | - Nora Kramkimel
- Department of Dermatology, Cochin University Hospital, AP-HP, Paris, France
| | - Sarah Guegan
- Department of Dermatology, Cochin University Hospital, AP-HP, Paris, France
| | - Selim Aractingi
- Department of Dermatology, Cochin University Hospital, AP-HP, Paris, France
| | - Olivier Huillard
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
| | - Jérôme Alexandre
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
- Université Paris Cité, INSERM, Centre de Recherche des Cordeliers, Équipe labélisée Ligue Contre le Cancer, CNRS SNC 5096, Sorbonne Université, Paris, France
| | - Marie Wislez
- Department of Pneumology, Cochin University Hospital, AP-HP, Paris, France
| | - François Goldwasser
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
- Université Paris Cité, Faculté de Médecine, INSERM, U1016, Institut Cochin, Paris, France
| | - Benoit Blanchet
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
- Université Paris Cité, Faculté de Pharmacie de Paris, UMR8038 CNRS CiTCoM, U1268 INSERM, CARPEM, Paris, France
| |
Collapse
|
5
|
Guo Y, Remaily BC, Thomas J, Kim K, Kulp SK, Mace TA, Ganesan LP, Owen DH, Coss CC, Phelps MA. Antibody Drug Clearance: An Underexplored Marker of Outcomes with Checkpoint Inhibitors. Clin Cancer Res 2024; 30:942-958. [PMID: 37921739 PMCID: PMC10922515 DOI: 10.1158/1078-0432.ccr-23-1683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/23/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Immune-checkpoint inhibitor (ICI) therapy has dramatically changed the clinical landscape for several cancers, and ICI use continues to expand across many cancer types. Low baseline clearance (CL) and/or a large reduction of CL during treatment correlates with better clinical response and longer survival. Similar phenomena have also been reported with other monoclonal antibodies (mAb) in cancer and other diseases, highlighting a characteristic of mAb clinical pharmacology that is potentially shared among various mAbs and diseases. Though tempting to attribute poor outcomes to low drug exposure and arguably low target engagement due to high CL, such speculation is not supported by the relatively flat exposure-response relationship of most ICIs, where a higher dose or exposure is not likely to provide additional benefit. Instead, an elevated and/or increasing CL could be a surrogate marker of the inherent resistant phenotype that cannot be reversed by maximizing drug exposure. The mechanisms connecting ICI clearance, therapeutic efficacy, and resistance are unclear and likely to be multifactorial. Therefore, to explore the potential of ICI CL as an early marker for efficacy, this review highlights the similarities and differences of CL characteristics and CL-response relationships for all FDA-approved ICIs, and we compare and contrast these to selected non-ICI mAbs. We also discuss underlying mechanisms that potentially link mAb CL with efficacy and highlight existing knowledge gaps and future directions where more clinical and preclinical investigations are warranted to clearly understand the value of baseline and/or time-varying CL in predicting response to ICI-based therapeutics.
Collapse
Affiliation(s)
- Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Bryan C. Remaily
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Justin Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Kyeongmin Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Samuel K. Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Latha P. Ganesan
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Dwight H. Owen
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Mitch A. Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| |
Collapse
|
6
|
Advances in pharmacokinetics and pharmacodynamics of PD-1/PD-L1 inhibitors. Int Immunopharmacol 2023; 115:109638. [PMID: 36587500 DOI: 10.1016/j.intimp.2022.109638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/09/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are a group of drugs designed to improve the therapeutic effects on various types of malignant tumors. Irrespective of monotherapy or combinational therapies as first-line and later-line therapy, ICIs have achieved benefits for various tumors. Programmed cell death protein-1 (PD-1) / ligand 1 (PD-L1) is an immune checkpoint that suppresses antitumor immunity, especially in the tumor microenvironment (TME). PD-1/PD-L1 immune checkpoint inhibitors block tumor-related downregulation of the immune system, thereby enhancing antitumor immunity. In comparison with traditional small-molecule drugs, ICIs exhibit pharmacokinetic characteristics owing to their high molecular weight. Furthermore, different types of ICIs exhibit different pharmacodynamic characteristics. Hence, ICIs have been approved for different indications by the Food and Drug Administration (FDA) and National Medical Products Administration (NMPA). This review summarizes pharmacokinetic and pharmacodynamic studies of PD-1/ PD-L1 inhibitors to provide a reference for rational clinical application.
Collapse
|
7
|
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
|
8
|
Liu SV, Nagasaka M, Stefaniak V, Gruver K, Lin Y, Ferry D, Socinski MA, Zhang L. The Applicability of the Results in the Asian Population of ORIENT-11 to a Western Population According to the ICH-E5 Framework. Front Oncol 2022; 12:859892. [PMID: 35756655 PMCID: PMC9226396 DOI: 10.3389/fonc.2022.859892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022] Open
Abstract
Sintilimab combined with pemetrexed and platinum met the primary endpoint of improving progression-free survival (PFS) as a first-line therapy for nonsquamous non-small cell lung cancer (NSCLC) in the phase 3 trial ORIENT-11 (NCT03607539). As seen in similar trials, the addition of sintilimab, a PD-1 inhibitor, to chemotherapy improved the PFS without significantly worsening the toxicity, with improvements in response rate and duration of response. In contrast to previous trials, the ORIENT-11 trial was conducted completely in China. Both intrinsic and extrinsic factors are important to consider when reviewing foreign clinical trial data, as they may influence the efficacy and the safety outcomes. Here we discuss the applicability of ORIENT-11 clinical results to a Western population.
Collapse
Affiliation(s)
- Stephen V Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
| | - Misako Nagasaka
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Irvine, CA, United States
| | | | - Kristi Gruver
- Oncology, Eli Lilly and Company, Indianapolis, IN, United States
| | - Yong Lin
- Oncology, Eli Lilly and Company, Indianapolis, IN, United States
| | - David Ferry
- Oncology, Eli Lilly and Company, Indianapolis, IN, United States
| | - Mark A Socinski
- Hematology and Oncology, AdventHealth Cancer Institute, Orlando, FL, United States
| | - Li Zhang
- Medical Oncology Department, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
9
|
Dynamics of Acquired Resistance to Nivolumab Therapies Varies From Administration Strategies. Clin Ther 2021; 43:2088-2103. [PMID: 34782163 DOI: 10.1016/j.clinthera.2021.10.004] [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: 04/08/2021] [Revised: 09/24/2021] [Accepted: 10/06/2021] [Indexed: 11/22/2022]
Abstract
PURPOSE The identification of optimal drug administration schedules to overcome the emergence of resistance that causes treatment failure is a major challenge in cancer research. We report the outcomes of a computational strategy to assess the dynamics of tumor progression as a function of time under different treatment regimens. METHODS We developed an evolutionary game theory model that combined Lotka-Volterra equations and pharmacokinetic properties with 2 competing cancer species: nivolumab-response cells and Janus kinase (JAK1/2) mutation cells. We selected 3 therapeutic schemes that have been tested in the clinical trials: 3 mg/kg Q2w, 10 mg/kg Q2w, and 480 mg Q4w. The simulation was performed under the intervals of 75, 125, and 175 days, respectively, for each regimen. The data sources of the pharmacokinetic parameters used in this study were collected from previous published clinical trials. Other parameters in the evolutionary model come from the existing references. FINDINGS Predictions under various dose schedules indicated a strong selection for nivolumab-independent cells. Under the 3 mg/kg dose strategy, the reproduction rate of JAK mutation cells was highest, with strongest tumor elimination ability at a 75-day interval between treatments. Prolonged drug intervals to 125 or 175 days delayed tumor evolution but accelerated tumor recurrence. Although 10 mg/kg Q2w had an obvious clinical effect in a short time, it further promotes the progress of resistant population compared with the 3 mg/kg dose. Our model suggests that 480 mg Q4w would be more valuable in terms of clinical efficacy, but complete resistant occurs earlier regardless the interval. IMPLICATIONS The results of this study emphasize that increasing the dose or shortening the interval between doses accelerates the evolution of heterogeneous populations, although the short-term effect is significant. In practice, the therapeutic regimen should be balanced according to the evolutionary principle.
Collapse
|
10
|
Zhang S, Gong C, Ruiz-Martinez A, Wang H, Davis-Marcisak E, Deshpande A, Popel AS, Fertig EJ. Integrating single cell sequencing with a spatial quantitative systems pharmacology model spQSP for personalized prediction of triple-negative breast cancer immunotherapy response. ACTA ACUST UNITED AC 2021; 1-2. [PMID: 34708216 PMCID: PMC8547770 DOI: 10.1016/j.immuno.2021.100002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Response to cancer immunotherapies depends on the complex and dynamic interactions between T cell recognition and killing of cancer cells that are counteracted through immunosuppressive pathways in the tumor microenvironment. Therefore, while measurements such as tumor mutational burden provide biomarkers to select patients for immunotherapy, they neither universally predict patient response nor implicate the mechanisms that underlie immunotherapy resistance. Recent advances in single-cell RNA sequencing technology measure cellular heterogeneity within cells of an individual tumor but have yet to realize the promise of predictive oncology. In addition to data, mechanistic multiscale computational models are developed to predict treatment response. Incorporating single-cell data from tumors to parameterize these computational models provides deeper insights into prediction of clinical outcome in individual patients. Here, we integrate whole-exome sequencing and scRNA-seq data from Triple-Negative Breast Cancer patients to model neoantigen burden in tumor cells as input to a spatial Quantitative System Pharmacology model. The model comprises a four-compartmental Quantitative System Pharmacology sub-model to represent a whole patient and a spatial agent-based sub-model to represent tumor volumes at the cellular scale. We use the high-throughput single-cell data to model the role of antigen burden and heterogeneity relative to the tumor microenvironment composition on predicted immunotherapy response. We demonstrate how this integrated modeling and single-cell analysis framework can be used to relate neoantigen heterogeneity to immunotherapy treatment outcomes. Our results demonstrate feasibility of merging single-cell data to initialize cell states in multiscale computational models such as the spQSP for personalized prediction of clinical outcomes to immunotherapy.
Collapse
Affiliation(s)
- Shuming Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chang Gong
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alvaro Ruiz-Martinez
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Emily Davis-Marcisak
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Atul Deshpande
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
11
|
Hata H, Mio T, Yamashita D, Matsumura C, Chisaki Y, Motohashi H, Yano Y. Factors Associated With Efficacy and Nivolumab-Related Interstitial Pneumonia in Non-Small Cell Lung Cancer: A Retrospective Survey. Cancer Control 2021; 27:1073274820977200. [PMID: 33297768 PMCID: PMC8480353 DOI: 10.1177/1073274820977200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Immune-checitors have been established as a novel standard treatment for non-small cell lung cancer (NSCLC). The aim of this study was to identify factors associated with efficacy and nivolumab-related interstitial pneumonia in NSCLC by evaluating clinical data at the initiation of and during treatment. METHODS We retrospectively reviewed the medical records of patients who underwent treatment with nivolumab between October 2015 and December 2017. Using pretreatment patient data, we investigated factors associated with overall survival (OS) and the onset of nivolumab-related pneumonitis. We investigated serum biochemistry during treatment to identify the determinants associated with progressive disease (PD) and the onset of nivolumab-related pneumonitis. RESULTS A total of 94 patients were included. Eleven patients continued treatment, and 54 patients were diagnosed with progressive disease. Nivolumab-related pneumonitis occurred in 15 patients. A pretreatment Eastern Cooperative Oncology Group Performance Status (ECOG PS) = 0 was linked to significantly longer OS than ECOG PS = 1 (median: 20.1 vs. 6.5 months, respectively; p < 0.001). There was a higher incidence of nivolumab-related pneumonitis in patients with a history of interstitial pneumonia than in those without it (p = 0.008). During treatment, the level of albumin gradually decreased prior to PD and onset of nivolumab-related pneumonitis. CONCLUSION These results suggest that the pretreatment ECOG PS is the determining factor that is associated with OS, whereas history of interstitial pneumonia is the factor associated with nivolumab-related pneumonitis. A decrease in albumin during treatment may be associated with both PD and nivolumab-related pneumonitis.
Collapse
Affiliation(s)
- Hiroki Hata
- Department of Pharmacy, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.,Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| | - Tadashi Mio
- Division of Respiratory Medicine, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Daisuke Yamashita
- Department of Pharmacy, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Chikako Matsumura
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| | - Yugo Chisaki
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| | - Hideyuki Motohashi
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan.,Department of Pharmaceutics, Osaka University of Pharmaceutical Sciences, Nasahara, Takatsuki, Osaka, Japan
| | - Yoshitaka Yano
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, Japan
| |
Collapse
|
12
|
Osarogiagbon RU, Sineshaw HM, Unger JM, Acuña-Villaorduña A, Goel S. Immune-Based Cancer Treatment: Addressing Disparities in Access and Outcomes. Am Soc Clin Oncol Educ Book 2021; 41:1-13. [PMID: 33830825 DOI: 10.1200/edbk_323523] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Avoidable differences in the care and outcomes of patients with cancer (i.e., cancer care disparities) emerge or worsen with discoveries of new, more effective approaches to cancer diagnosis and treatment. The rapidly expanding use of immunotherapy for many different cancers across the spectrum from late to early stages has, predictably, been followed by emerging evidence of disparities in access to these highly effective but expensive treatments. The danger that these new treatments will further widen preexisting cancer care and outcome disparities requires urgent corrective intervention. Using a multilevel etiologic framework that categorizes the targets of intervention at the individual, provider, health care system, and social policy levels, we discuss options for a comprehensive approach to prevent and, where necessary, eliminate disparities in access to the clinical trials that are defining the optimal use of immunotherapy for cancer, as well as its safe use in routine care among appropriately diverse populations. We make the case that, contrary to the traditional focus on the individual level in descriptive reports of health care disparities, there is sequentially greater leverage at the provider, health care system, and social policy levels to overcome the challenge of cancer care and outcomes disparities, including access to immunotherapy. We also cite examples of effective government-sponsored and policy-level interventions, such as the National Cancer Institute Minority-Underserved Community Oncology Research Program and the Affordable Care Act, that have expanded clinical trial access and access to high-quality cancer care in general.
Collapse
Affiliation(s)
| | | | - Joseph M Unger
- Health Services Research, Public Health Sciences Division, Fred Hutchinson Cancer Research Center Affiliate, University of Washington, Seattle, WA
| | | | - Sanjay Goel
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
13
|
Sheng J, Zhang J, Baudelet C, Roy A. Clinical Benefit-Risk Assessment of Nivolumab 240 mg Every 2 Weeks in Chinese Patients With Advanced and Metastatic Solid Tumors. J Clin Pharmacol 2021; 61:1045-1053. [PMID: 33501654 PMCID: PMC8359491 DOI: 10.1002/jcph.1821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/21/2021] [Indexed: 12/11/2022]
Abstract
Nivolumab 240 mg every 2 weeks is approved in China by the National Medical Product Agency for squamous cell carcinoma of the head and neck and gastric cancer, based on population pharmacokinetic (PPK) analyses and benefit-risk assessment of safety/efficacy in solid tumors, including Chinese and global populations. The aim of this assessment was to investigate exposure and risk for adverse events (AEs) with flat dosing compared with weight-based dosing. Nivolumab 240-mg and 3-mg/kg every-2-week exposures in Chinese patients were simulated using PPK modeling, and AEs in Chinese and pooled global populations were compared by dosing regimen, exposure, and weight. The 10-mg/kg every-2-week regimen was included because it is known to be well tolerated. Predicted nivolumab exposure in Chinese patients receiving 240 mg every 2 weeks was ∼25% higher versus 3 mg/kg every 2 weeks, but ∼60% lower versus 10 mg/kg every 2 weeks. Grade 3/4 AE incidence in Chinese patients receiving nivolumab 3 mg/kg every 2 weeks was similar with 240-mg every-2-week dosing and with patients from global populations treated with 3 or 10 mg/kg every 2 weeks. There was no trend toward increased AE incidence with high versus low nivolumab exposure or in global patients of varying body weight receiving 3 or 10 mg/kg every 2 weeks. Objective response rates were similar in Chinese and global patients with squamous and nonsquamous NSCLC. Results showed that benefit-risk profiles with nivolumab 240 mg every 2 weeks were similar to those of the 3-mg/kg every-2-week regimen in Chinese patients and global populations, providing an alternative treatment option to Chinese patients.
Collapse
Affiliation(s)
| | - Jason Zhang
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | | | - Amit Roy
- Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| |
Collapse
|
14
|
Nishio M, Sugawara S, Atagi S, Akamatsu H, Sakai H, Okamoto I, Takayama K, Hayashi H, Nakagawa Y, Kawakami T. Subgroup Analysis of Japanese Patients in a Phase III Study of Atezolizumab in Extensive-stage Small-cell Lung Cancer (IMpower133). Clin Lung Cancer 2019; 20:469-476.e1. [PMID: 31466854 DOI: 10.1016/j.cllc.2019.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/20/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Atezolizumab is effective and well-tolerated in patients with extensive-stage small-cell lung cancer (ES-SCLC), but differences in response to systemic therapy exist between Asian and Caucasian patients. Here, we assess the efficacy and tolerability of atezolizumab in Japanese patients from the IMpower133 trial (NCT02763579). PATIENTS AND METHODS Key eligibility criteria for this multicenter, double-blind, placebo-controlled, randomized study included age ≥ 18 years; histologically or cytologically confirmed ES-SCLC, measurable per Response Evaluation Criteria in Solid Tumors version 1.1; an Eastern Cooperative Oncology Group performance status of 0/1; and no prior systemic treatment for ES-SCLC. Patients were treated with either atezolizumab 1200 mg or placebo with carboplatin (area under the curve of 5 mg/mL/min) and etoposide (100 mg/m2). Primary endpoints were overall survival and investigator-assessed progression-free survival in the intention-to-treat population. Of the 403 patients randomized in the IMpower133 trial, 42 were enrolled at Japanese centers. RESULTS In Japanese patients in the intention-to-treat population, the median overall survival in the atezolizumab group (n = 20) was longer than that in the placebo group (n = 22; 14.6 months; 95% confidence interval [CI], 11.8-17.8 months vs. 11.9 months; 95% CI, 8.4-15.8, respectively; hazard ratio, 0.72; 95% CI, 0.31-1.67). The median progression-free survival was 4.5 months (95% CI, 4.2-8.1 months) versus 4.0 months (95% CI, 2.9-5.6 months; hazard ratio, 0.47; 95% CI, 0.23-0.96), respectively. Atezolizumab was generally well-tolerated, with no treatment-related deaths. CONCLUSION The addition of atezolizumab to carboplatin and etoposide was effective and well-tolerated in Japanese patients with ES-SCLC. Results are consistent with the primary analysis of the IMpower133 trial.
Collapse
Affiliation(s)
- Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Miyagi, Japan
| | - Shinji Atagi
- Department of Thoracic Oncology, National Hospital Organization Kinki-chuo Chest Medical Center, Osaka, Japan
| | - Hiroaki Akamatsu
- Department of Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | - Hiroshi Sakai
- Department of Thoracic Oncology, Saitama Cancer Center, Saitama, Japan
| | - Isamu Okamoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | | | | |
Collapse
|