1
|
Tan Z, Völler S, Sancho-Araiz A, Knibbe CAJ, Moes DJAR. A Systematic Evaluation of the Dosing Regimens for Approved Targeted Therapies and Immune Checkpoint Inhibitors in Metastatic Renal Cell Carcinoma From a Project OPTIMUS Perspective. J Clin Pharmacol 2025. [PMID: 40313197 DOI: 10.1002/jcph.70035] [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: 02/10/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025]
Abstract
Targeted therapies and immune checkpoint inhibitors (ICIs) have significantly improved survival outcomes in metastatic renal cell carcinoma (mRCC) but are often associated with high rates of adverse events, leading to dose reductions or treatment discontinuation. The FDA's recent initiative, Project OPTIMUS, emphasizes the importance of optimizing dosing regimens in oncology clinical development, and moves beyond the conventional maximum tolerated dose approach. In this study, we aimed to review and redefine the approved dosing strategies for targeted therapies and ICIs in mRCC from the Project OPTIMUS perspective, including pazopanib, axitinib, cabozantinib, sunitinib, everolimus, and nivolumab. A comprehensive summary of FDA clinical pharmacology reviews and clinical studies performed in routine clinical practice was conducted, alongside model-informed simulations of pharmacokinetic profiles with approved and alternative regimens. Results demonstrated that actual tolerated doses in clinical practice were 46.1% to 86% lower than the approved dosages, with up to 75% of patients requiring dose adjustments. Model-informed simulations suggested that for most targeted therapies, a 14%-50% dose reduction maintained comparable efficacy while improving tolerability. For nivolumab, simulations confirmed adequate drug exposure with the approved flat dose regimens, without an increase of adverse effects. In conclusion, we identified optimized dosing regimens that could improve drug tolerability while maintaining efficacy for approved targeted therapies and ICIs in mRCC. We suggest that these optimized dosing regimens should be considered for use in clinical practice and that the optimal exposure range be included in drug labels to support pharmacokinetically guided dose individualization in clinical practice.
Collapse
Affiliation(s)
- Zhiyuan Tan
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Swantje Völler
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Aymara Sancho-Araiz
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Catherijne A J Knibbe
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Corridore S, Verreault M, Martin H, Delobel T, Carrère C, Idbaih A, Ballesta A. Circumventing glioblastoma resistance to temozolomide through optimal drug combinations designed by systems pharmacology and machine learning. Br J Pharmacol 2025. [PMID: 40229949 DOI: 10.1111/bph.70027] [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: 08/27/2024] [Revised: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND AND PURPOSE Glioblastoma (GBM), the most frequent and aggressive brain tumour in adults, is associated with a dismal prognostic despite intensive treatment involving surgery, radiotherapy and temozolomide (TMZ)-based chemotherapy. The initial or acquired resistance of GBM to TMZ appeals for precision medicine approaches to the design of novel efficient combination pharmacotherapies. Such investigation needs to account for the overexpression of the O6-methylguanine-DNA methyl-transferase (MGMT) repair enzyme which is responsible for TMZ resistance in patients. EXPERIMENTAL APPROACH A comprehensive approach combining quantitative systems pharmacology (QSP) models and machine learning (ML) was undertaken to design TMZ-based drug combinations circumventing the initial resistance to the alkylating agent. KEY RESULTS A QSP model representing TMZ cellular pharmacokinetics-pharmacodynamics and dysregulated pathways in GBM was developed and validated using multi-type time- and dose-resolved datasets, available in control or MGMT-overexpressing cells. In silico drug screening and subsequent experimental validation identified a strategy to re-sensitise TMZ-resistant cells consisting in combining TMZ with inhibitors of the base excision repair and of homologous recombination. Using ML, functional signatures of response to such optimal multi-agent therapy were derived to assist decision-making in patients. CONCLUSION AND IMPLICATIONS We successfully demonstrated the relevance of combined QSP and ML to design efficient drug combinations re-sensitising glioblastoma cells initially resistant to TMZ. The developed framework may further serve to identify personalised therapies and administration schedules by extending it to account for additional patient-specific altered pathways and whole-body features.
Collapse
Affiliation(s)
- Sergio Corridore
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
| | - Maïté Verreault
- AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, Paris, France
| | - Hugo Martin
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
- University of Rennes, EHESP, CNRS, Inserm, Arènes - UMR 6051, RSMS - U 1309, Rennes, France
| | - Thibault Delobel
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
| | - Cécile Carrère
- Institut Denis Poisson, Université d'Orléans, CNRS, Orléans, France
| | - Ahmed Idbaih
- AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, Sorbonne Université, Paris, France
| | - Annabelle Ballesta
- INSERM Unit 1331, Institut Curie, PSL Research University, CBIO-Center for Computational Biology, Mines Paris, Cancer Systems Pharmacology team, Saint Cloud, France
| |
Collapse
|
3
|
Demin O, Ou Y, Kolesova G, Shchelokov D, Stepanov A, Musatova V, Sahasranaman S, Zhao Y, Liu X, Tang Z, Hanley WD. Quantitative Systems Pharmacology Model to Predict Target Occupancy by Bruton Tyrosine Kinase Inhibitors in Patients With B-Cell Malignancies. CPT Pharmacometrics Syst Pharmacol 2025; 14:706-717. [PMID: 39937656 PMCID: PMC12001278 DOI: 10.1002/psp4.13307] [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: 10/09/2024] [Revised: 12/09/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025] Open
Abstract
The effectiveness of Bruton tyrosine kinase (BTK) inhibitors is influenced by the level of BTK occupancy in target tissues. In randomized phase 3 studies, progression-free survival (PFS) with zanubrutinib was superior to ibrutinib, whereas acalabrutinib was noninferior to ibrutinib in previously treated chronic lymphocytic leukemia. To establish a link between numerical differences in BTK occupancy and differentiated efficacy profiles among three covalent BTK inhibitors, quantitative systems pharmacology (QSP) modeling was employed. The model was developed to describe available clinical BTK occupancy data in patients with B-cell malignancies. Simulations of BTK occupancy were conducted for various clinical scenarios (e.g., dose interruption) and for bone marrow (BM), for which routine measurements are difficult. This model describes pharmacokinetics of BTK inhibitors; intracellular concentration of BTK inhibitors in peripheral blood mononuclear cells (PBMCs), BM, and lymph nodes (LNs); binding of BTK inhibitors with BTK; and BTK turnover rate. The model was validated using available clinical BTK occupancy data. Consistent with observed clinical data, the model predicted that zanubrutinib 160 mg twice daily resulted in higher median trough BTK occupancy in PBMCs, LNs, and BM compared with ibrutinib and acalabrutinib. Although the BTK occupancy differences at trough were relatively small between the BTK inhibitors, the differences were more pronounced after dose interruption. The current work underscores the importance of maintaining high BTK occupancy at steady-state trough and during treatment interruption to ensure maximal efficacy and provides an example of combining in vitro and clinical data to model receptor occupancy in tissues where measurements are challenging.
Collapse
Affiliation(s)
| | - Ying Ou
- Clinical PharmacologyBeiGene USA Inc.San MateoCaliforniaUSA
| | | | | | | | | | | | - Yating Zhao
- Clinical PharmacologyBeiGene USA Inc.San MateoCaliforniaUSA
| | - Xiangyu Liu
- Clinical PharmacologyBeiGene USA Inc.San MateoCaliforniaUSA
| | - Zhiyu Tang
- Clinical PharmacologyBeiGene USA Inc.San MateoCaliforniaUSA
| | | |
Collapse
|
4
|
Teruel CF, Cullberg M, González-García I, Schiavon G, Zhou D. An exposure-safety analysis to support the dosage of the novel AKT inhibitor capivasertib. Cancer Chemother Pharmacol 2025; 95:48. [PMID: 40153000 PMCID: PMC11953117 DOI: 10.1007/s00280-025-04775-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/18/2025] [Indexed: 03/30/2025]
Abstract
PURPOSE This study aimed to evaluate capivasertib exposure-response relationships for clinical safety events to support dosage selection. METHODS Data from 277 patients with solid tumors participating in three phase 1 studies were analyzed. Capivasertib 80-800 mg was administered as monotherapy orally twice daily (BID) on continuous or intermittent (4 days on, 3 days off [4/3] or 2 days on, 5 days off [2/5]) schedules. Relationships between exposure related metrics (dose, weekly dose, AUC, AUCPWD, Cmax, and Cmin) and probability of safety endpoints (adverse event [AE] leading to dose discontinuation, AE leading to dose modification, serious AE [SAE], AE grade ≥ 3, AE grade ≥ 1, diarrhea AE grade ≥ 2, rash AE grade ≥ 2, hyperglycemia AE grade ≥ 3 and increased blood glucose > 13.9 mmol/L) were evaluated by logistic regression. RESULTS Significant exposure-response relationships were identified for all safety endpoints evaluated, except for AE grade ≥ 1. The analysis suggested that most of the safety endpoints are driven by the total weekly exposure, whereas glucose elevations are driven by the exposure achieved within a dosing interval. The probability of experiencing an AE leading to dose discontinuation, AE leading to dose modification, SAE, AE grade ≥ 3, diarrhea or rash were lower with the 480 mg BID [4/3] schedule than with the 320 mg BID continuous schedule. CONCLUSION Significant exposure-response relationships were identified for safety endpoints when capivasertib was administered to patients with solid tumors suggesting that the intermittent [4/3] schedule is better tolerated than the continuous schedule due to lower total weekly exposure.
Collapse
Affiliation(s)
- Carlos Fernandez Teruel
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Marie Cullberg
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ignacio González-García
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Gaia Schiavon
- Late Development Oncology, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Diansong Zhou
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA.
| |
Collapse
|
5
|
Remon J, Bortolot M, Bironzo P, Cortiula F, Menis J, Brandao M, Naidoo J, van Geel R, Reguart N, Arrieta O, Mountzios G, Hendriks LEL, Besse B. De-Escalation Strategies With Immune Checkpoint Blockers in Non-Small Cell Lung Cancer: Do We Already Have Enough Evidence? J Clin Oncol 2025; 43:1148-1156. [PMID: 39836933 DOI: 10.1200/jco-24-02347] [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: 10/20/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 01/23/2025] Open
Abstract
Immune checkpoint blockers (ICBs) have revolutionized the treatment of non-small cell lung cancer (NSCLC). Currently, one-dose-fits-all maximalist regimens have been considered the standard of care, with ICBs administered at flat doses regardless of patients' weight. Treatment duration with ICBs is often arbitrary across stages, ranging from a fixed time point to until disease progression or unacceptable toxicity. However, the pharmacokinetic and pharmacodynamic properties of ICBs differ significantly from those of traditional cytotoxic drugs and the approved and selected doses on the basis of the maximum tolerated dose are often overestimated as there is limited evidence supporting a direct relationship between therapeutic intensity and outcomes. This can lead to overtreatment of patients, resulting in an increased risk of toxicity without enhanced efficacy. In addition, the use of these drugs is associated with significant costs that burden the global health care system and exacerbate disparities in access to care. De-escalating treatment by reducing the dose, duration, and frequency of administration of ICBs could optimize treatment efficacy, reduce toxicities, improve patients' quality of life, and even decrease costs. Ultimately, de-escalation strategies may help to reduce treatment inequalities and to improve drug access worldwide. The aim of this review is to summarize and discuss the main issues and challenges regarding the de-escalation of ICBs in patients with NSCLC, focusing on dose-intensity reduction and treatment duration selection. Moreover, we assess the economic impact of implementing de-escalation approaches.
Collapse
Affiliation(s)
- Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Martina Bortolot
- Department of Pulmonary Diseases, Maastricht University Medical Centre+, GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Paolo Bironzo
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of Oncology, University of Turin, Turin, Italy
| | - Francesco Cortiula
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre (+), GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
- University Hospital of Udine, Department of Oncology, Udine, Italy
| | - Jessica Menis
- Medical Oncology Department, University and Hospital Trust of Verona, Verona, Italy
| | - Mariana Brandao
- Institute Jules Bordet-Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | - Jarushka Naidoo
- Beaumont Hospital and RCSI University of Health Sciences, Dublin, Ireland
| | - Robin van Geel
- Department of Clinical Pharmacy & Toxicology, Maastricht University Medical Centre+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Noemi Reguart
- Department of Oncology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, Maastricht University Medical Centre+, GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
6
|
Zhao Y, Tsujimoto A, Ide T, Zhang J, Feng Y, Gao L, Bello A, Roy A. Model-based population pharmacokinetic and exposure response analyses for safety and efficacy of nivolumab as adjuvant treatment in subjects with resected oesophageal or gastroesophageal junction cancer. Br J Clin Pharmacol 2024; 90:2920-2930. [PMID: 39054780 DOI: 10.1111/bcp.16188] [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: 12/06/2023] [Revised: 06/09/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
AIMS Nivolumab is approved as adjuvant treatment in subjects with resected oesophageal or gastroesophageal junction cancer (EC/GEJC) based on results from the pivotal CheckMate 577 trial. We present a model-based clinical pharmacology profiling and benefit-risk assessment of nivolumab as adjuvant treatment in subjects with resected EC/GEJC supporting a less frequent dosing regimen. METHODS Population pharmacokinetic (popPK) analysis was conducted to characterize nivolumab pharmacokinetics (PK) using clinical data from 1493 subjects from seven monotherapy clinical studies across multiple solid tumours. The exposure-response (E-R) analyses included data from 756 patients from CheckMate 577. E-R relationships for efficacy and safety were characterized by evaluating the relationship between nivolumab exposure and disease-free survival (DFS) for efficacy; and time to first occurrence of Grade ≥2 immune-mediated adverse events (Gr2 + IMAEs) for safety. RESULTS Nivolumab exposure was found to be associated with both DFS and risk of Gr2 + IMAEs. However, the hazard ratios (HRs) (95% confidence interval [CI]) at the 5th and 95th percentiles of nivolumab exposure were similar for DFS and Gr2 + IMAEs, indicating flat E-R relationships within the exposure range produced by the studied regimen. Model-predicted probability of DFS and Gr2 + IMAEs were similar between the two regimens of 240 mg every 2 weeks or 480 mg every 4 weeks for 16 weeks followed by 480 mg Q4W up to 1 year. CONCLUSIONS The analyses demonstrated a flat E-R relationship over the range of exposures produced by the studied regimen and supported the approval of an alternative dosing regimen with less frequent dosing in patients with adjuvant EC/GEJC.
Collapse
Affiliation(s)
- Yue Zhao
- Bristol Myers Squibb, Princeton, NJ, United States
| | | | - Takafumi Ide
- Bristol Myers Squibb, Princeton, NJ, United States
| | - Jenny Zhang
- Bristol Myers Squibb, Princeton, NJ, United States
| | - Yan Feng
- Bristol Myers Squibb, Princeton, NJ, United States
| | - Ling Gao
- Bristol Myers Squibb, Princeton, NJ, United States
| | | | - Amit Roy
- Bristol Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
7
|
Cui C, Wang J, Wang C, Xu T, Qin L, Xiao S, Gong J, Song L, Liu D. Model-informed drug development of envafolimab, a subcutaneously injectable PD-L1 antibody, in patients with advanced solid tumors. Oncologist 2024; 29:e1189-e1200. [PMID: 38982653 PMCID: PMC11379657 DOI: 10.1093/oncolo/oyae102] [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: 11/07/2023] [Accepted: 04/17/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Envafolimab is the first and only globally approved subcutaneously injectable PD-L1 antibody for the treatment of instability-high (MSI-H) or DNA mismatch repair deficient (dMMR) advanced solid tumors in adults, including those with advanced colorectal cancer that has progressed after treatment with a fluoropyrimidine, oxaliplatin, and irinotecan. The aim of this investigation was to examine the pharmacokinetic and exposure-response (E-R) profile of envafolimab in patients with solid tumors to support the approval of fixed and alternative dose regimens. METHODS In this study, a population pharmacokinetic (PopPK) modeling approach will be employed to quantitatively evaluate intrinsic and extrinsic covariates. Additionally, PopPK-estimated exposure parameters were used to evaluate E-R relationship for safety and efficacy to provide a theoretical basis for recommending optimal treatment regimens. Simulations were performed on the dosing regimens of body weight-based regimen of 2.50 mg/kg QW, fixed dose 150 mg QW, and 300 mg Q2W for the selection of alternative dosing regimens. Data from 4 clinical studies (NCT02827968, NCT03101488, NCT03248843, and NCT03667170) were utilized. RESULTS The PopPK dataset comprised 182 patients with 1810 evaluable envafolimab concentration records. Finally, a one-compartment model incorporating first-order absorption, first-order linear elimination, and time-dependent elimination according to an Emax function was found to accurately describe the concentration-time data of envafolimab in patients with advanced solid tumors. Creatinine clearance and country were identified as statistically significant factors affecting clearance, but had limited clinical significance. A relative flat exposure-response relationship was observed between early measures of safety and efficacy to verify that no dose adjustment is required. Simulation results indicated that 2.50 mg/kg QW, 150 mg QW, and 300 mg Q2W regimen yield similar steady-state exposure. CONCLUSIONS No statistically significant difference was observed between weight-based and fixed dose regimens. Model-based simulation supports the adoption of a 150 mg weekly or 300 mg biweekly dosing regimen of envafolimab in the solid tumor population, as these schedules effectively balance survival benefits and safety risks.
Collapse
Affiliation(s)
- Cheng Cui
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Jing Wang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Chunyang Wang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Ting Xu
- Alphamab Co., Ltd., Suzhou, People’s Republic of China
| | - Lan Qin
- 3DMedicines Co., Ltd., Shanghai, People’s Republic of China
| | - Shen Xiao
- 3DMedicines Co., Ltd., Shanghai, People’s Republic of China
| | - John Gong
- 3DMedicines Co., Ltd., Shanghai, People’s Republic of China
| | - Ling Song
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, People’s Republic of China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Truong J, Yeung SST, Kletas V, de Lemos M, Schaff K, Nakashima L. Utilization and toxicity patterns of 2-weekly (Q2W) versus 4-weekly (Q4W) nivolumab for treatment of adjuvant and metastatic melanoma at BC cancer. J Oncol Pharm Pract 2024; 30:1016-1022. [PMID: 37654194 DOI: 10.1177/10781552231199048] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
BACKGROUND Nivolumab, an immune checkpoint inhibitor used to treat several malignancies, is associated with immune-related adverse events (IrAEs). Original dosing for melanoma was 3 mg/kg (maximum 240 mg) every 2 weeks (Q2W). Based on simulation studies depicting similar efficacy and toxicity to original dosing, extended interval dosing of 6 mg/kg (maximum 480 mg) every 4 weeks (Q4W) was introduced. OBJECTIVE This study will compare safety between Q2W and Q4W dosing at BC Cancer in melanoma patients. METHODS Retrospective chart review for reported incidence, onset, and severity of IrAEs in melanoma patients treated with nivolumab Q2W and Q4W dosing was completed. Fisher's test was conducted for first incidence IrAEs using Microsoft Excel. RESULTS Seventy-one patients were identified (Q2W n = 35, Q4W n = 36). Baseline characteristics were similar in both groups. No statistically significant difference was found in incidence of IrAEs between Q2W and Q4W dosing (Q2W 40% vs Q4W 50%, p = 0.477). Rash was most common (Q2W 79% vs Q4W 50%) followed by hypothyroidism (Q2W 33% vs Q4W 20%). Median onset of IrAEs seemed later with Q4W dosing (Q2W cycle 1 vs Q4W cycle 4). Regardless of dosing, most IrAEs were grade 1-2 in severity (Q2W 100% vs Q4W 89%). CONCLUSION Q4W dosing is associated with comparable incidence and potentially later onset of IrAEs compared to Q2W dosing. Most IrAEs in both dosing groups were similar and mild. Therefore, Q4W dosing offers a safe alternative to Q2W dosing while providing benefits including decreased workload for staff, decreased clinic visits, and viral exposure by patients.
Collapse
|
9
|
Schenker M, Burotto M, Richardet M, Ciuleanu TE, Gonçalves A, Steeghs N, Schoffski P, Ascierto PA, Maio M, Lugowska I, Lupinacci L, Leary A, Delord JP, Grasselli J, Tan DSP, Friedmann J, Vuky J, Tschaika M, Konduru S, Vemula SV, Slepetis R, Kollia G, Pacius M, Duong Q, Huang N, Doshi P, Baden J, Di Nicola M. Randomized, open-label, phase 2 study of nivolumab plus ipilimumab or nivolumab monotherapy in patients with advanced or metastatic solid tumors of high tumor mutational burden. J Immunother Cancer 2024; 12:e008872. [PMID: 39107131 PMCID: PMC11308901 DOI: 10.1136/jitc-2024-008872] [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] [Accepted: 06/14/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND Checkpoint inhibitor therapy has demonstrated overall survival benefit in multiple tumor types. Tumor mutational burden (TMB) is a predictive biomarker for response to immunotherapies. This study evaluated the efficacy of nivolumab+ipilimumab in multiple tumor types based on TMB status evaluated using either tumor tissue (tTMB) or circulating tumor DNA in the blood (bTMB). PATIENTS AND METHODS Patients with metastatic or unresectable solid tumors with high (≥10 mutations per megabase) tTMB (tTMB-H) and/or bTMB (bTMB-H) who were refractory to standard therapies were randomized 2:1 to receive nivolumab+ipilimumab or nivolumab monotherapy in an open-label, phase 2 study (CheckMate 848; NCT03668119). tTMB and bTMB were determined by the Foundation Medicine FoundationOne® CDx test and bTMB Clinical Trial Assay, respectively. The dual primary endpoints were objective response rate (ORR) in patients with tTMB-H and/or bTMB-H tumors treated with nivolumab+ipilimumab. RESULTS In total, 201 patients refractory to standard therapies were randomized: 135 had tTMB-H and 125 had bTMB-H; 82 patients had dual tTMB-H/bTMB-H. In patients with tTMB-H, ORR was 38.6% (95% CI 28.4% to 49.6%) with nivolumab+ipilimumab and 29.8% (95% CI 17.3% to 44.9%) with nivolumab monotherapy. In patients with bTMB-H, ORR was 22.5% (95% CI 13.9% to 33.2%) with nivolumab+ipilimumab and 15.6% (95% CI 6.5% to 29.5%) with nivolumab monotherapy. Early and durable responses to treatment with nivolumab+ipilimumab were seen in patients with tTMB-H or bTMB-H. The safety profile of nivolumab+ipilimumab was manageable, with no new safety signals. CONCLUSIONS Patients with metastatic or unresectable solid tumors with TMB-H, as determined by tissue biopsy or by blood sample when tissue biopsy is unavailable, who have no other treatment options, may benefit from nivolumab+ipilimumab. TRIAL REGISTRATION NUMBER NCT03668119.
Collapse
Affiliation(s)
- Michael Schenker
- Sf Nectarie Oncology Center and University of Medicine and Pharmacy, Craiova, Romania
| | | | - Martin Richardet
- Fundación Richardet Longo, Instituto Oncológico de Córdoba, Córdoba, Argentina
| | - Tudor-Eliade Ciuleanu
- Department of Oncology, Oncology Institute Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
- Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anthony Gonçalves
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Patrick Schoffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Paolo A Ascierto
- Department of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Michele Maio
- Department of Oncology, University of Siena and Center for Immuno-Oncology, Siena, Italy
| | - Iwona Lugowska
- Department of Early Phase Clinical Trials, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Alexandra Leary
- Université Paris-Saclay and Institut Gustave‑Roussy, Villejuif, France
| | - Jean-Pierre Delord
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Toulouse, France
| | - Julieta Grasselli
- Center for Medical Education and Clinical Research (CEMIC) University Hospital, Buenos Aires, Argentina
| | - David S P Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
- Cancer Science Institute, National University of Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore
| | - Jennifer Friedmann
- Segal Cancer Center, Jewish General Hospital, Montreal, Québec, Canada
- Rossy Cancer Network, McGill University, Montreal, Québec, Canada
| | - Jacqueline Vuky
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | | | | | - Quyen Duong
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Ning Huang
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Parul Doshi
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Massimo Di Nicola
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
10
|
Tasaki Y, Ito N, Mimura Y, Sugiyama Y, Ogawa R, Shimura T, Nakamura M, Kawakita D, Hamamoto S, Uemura T, Yokota K, Iida M, Odagiri K, Kimura Y, Hotta Y, Komatsu H, Okuda K, Niimi A, Yasui T, Iwasaki S, Morita A, Kataoka H, Takiguchi S, Furukawa-Hibi Y. Real-world data on efficacy/safety and economic impact of nivolumab administered every 2 and 4 weeks among Japanese patients. Asia Pac J Clin Oncol 2024; 20:515-521. [PMID: 38682421 DOI: 10.1111/ajco.14073] [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: 11/25/2023] [Revised: 03/06/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
AIM A new treatment interval for nivolumab administration at 480 mg every 4 weeks, in addition to 240 mg every 2 weeks, was approved in Japan in 2020. Using model-based evaluation, it was speculated that the effects or safety of nivolumab do not differ between the two treatment intervals; however, real-world data on nivolumab efficacy, safety, and economic impact are lacking. Accordingly, we aimed to examine the effects of nivolumab treatment intervals (2 weeks vs. 4 weeks) in terms of efficacy, safety, and economic impact in Japanese patients with cancer. METHODS We retrospectively analyzed 126 patients treated with nivolumab. The patients were divided into two groups depending on whether they received nivolumab at 240 mg every 2 weeks (2-week group) or 480 mg every 4 weeks (4-week group). RESULTS Efficacy results found no significant difference between the 4- and 2-week groups considering median overall survival (p = 0.70) and median progression-free survival (p = 0.57). The incidence of any grade and ≥ grade 3 immune-related adverse events did not differ between the 4-week and 2-week groups (any grade, p = 0.13; ≥ grade 3, p = 0.36). Excluding drug costs, the 4-week group had significantly lower medical costs than the 2-week group (2-week vs. 4-week: mean, 94,659 JPY [679.0 USD] vs. 58,737 JPY [421.3 USD]; p < 0.05). CONCLUSION Collectively, our findings suggest that nivolumab 480 mg every 4 weeks may be more effective than nivolumab 240 mg every 2 weeks in terms of economic impact.
Collapse
Affiliation(s)
- Yoshihiko Tasaki
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Nanami Ito
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshihisa Mimura
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yosuke Sugiyama
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Motoki Nakamura
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Daisuke Kawakita
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takehiro Uemura
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Keisuke Yokota
- Department of Thoracic and Pediatric Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Moeko Iida
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kunihiro Odagiri
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuka Kimura
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuji Hotta
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirokazu Komatsu
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Katsuhiro Okuda
- Department of Thoracic and Pediatric Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy, and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shinichi Iwasaki
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoko Furukawa-Hibi
- Department of Clinical Pharmaceutics, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
11
|
Elijah J, Puzanov I, Cresanti B, Hamad L, Attwood K, Catalfamo K, Riebandt G. Evaluation of safety outcomes between nivolumab regimens with differing dosing patterns. J Oncol Pharm Pract 2024:10781552241264817. [PMID: 39043219 DOI: 10.1177/10781552241264817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
BACKGROUND Real-world safety outcomes between the two flat-dose nivolumab regimens demonstrated to be similar in a study of adjuvant nivolumab recipients for melanoma. However, this study was limited by a single oncology patient population, a small sample size, and insufficient study power. The primary objective of this study was to evaluate the incidence of immunotherapy-related adverse effects (irAEs) between nivolumab regimens with differing dosing patterns in various solid tumor patient populations. METHODS Single-center retrospective cohort study of adult patients with solid tumor malignancies who received nivolumab 240 mg Q2W or 480 mg Q4W, or who were transitioned from 240 mg Q2W to 480 mg Q4W from March 1, 2018 to March 31, 2022 were selected for analysis from an electronic health record generated report. The primary endpoint evaluated was the incidence of irAEs. Secondary endpoints included the incidence of significant irAEs and reasons for treatment discontinuation. These endpoints were compared by univariate analysis between all three cohorts. A multivariate analysis was then conducted for the primary endpoint. RESULTS Nivolumab 240 mg Q2W was associated with a statistically significant increase in the incidence of colitis whereas the 480 mg Q4W regimen was associated with a statistically significant increase in the incidence of pruritis. The incidence of irAEs was not different between the three cohorts, while the incidence of significant irAEs was higher in the 240 mg Q2W and 240 mg Q2W to 480 mg Q4W cohorts. CONCLUSION Clinicians ought to be aware of differences in the irAE profiles between nivolumab regimens with differing dosing patterns.
Collapse
Affiliation(s)
- Joseph Elijah
- School of Pharmacy and Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Igor Puzanov
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Lamya Hamad
- Department of Clinical Pharmacy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kayla Catalfamo
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Grazyna Riebandt
- Department of Clinical Pharmacy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
12
|
Leroy M, Desmedt E, Deramoudt L, Vasseur M, Odou P, Béhal H, Décaudin B, Mortier L, Simon N. Retrospective comparison of a weight-based dose every 2 weeks with a fixed dose every month: a real-life analysis of nivolumab in the treatment of advanced melanoma. Melanoma Res 2024; 34:258-264. [PMID: 38489575 DOI: 10.1097/cmr.0000000000000965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Nivolumab was first authorized at a weight-based dose (WBD) of 3 mg/kg every two weeks (Q2W). Since 2017, a fixed dose (FD) regimen [first 240 mg Q2W and then 480 mg per month (Q4W)] was allowed. The objective of the study was to compare a WBD regimen and an FD regimen with regard to effectiveness and safety. We conducted a single-center, retrospective, real-life study of consecutive adult patients who had received a WBD of nivolumab or an FD of 480 mg Q4W. The primary endpoint was the occurrence of grade ≥3 immune-related adverse events (irAEs). The secondary endpoints were overall survival and cost of the treatment. In all, 342 patients were included: 71 in the WBD cohort and 271 in the FD cohort. Of these patients, 201 patients (59.6%) experienced an irAE, and 24 of these events were graded as ≥3. At 12 months, there was no significant difference in irAE occurrence between the two cohorts [hazard ratio (95% confidence interval): 0.54 (0.21-1.36), P = 0.19]. The 12-month overall survival rate was significantly lower in the WBD cohort ( P < 0.001). Switching from a fortnightly weight dose to a fixed monthly dose halves the cost of hospitalization. Our results did not show a significant difference between WBD and FD cohort in the occurrence of severe irAEs. However overall survival appeared to be significantly higher in FD group. Some clinical trials are investigating a hybrid dosing regimen in which a WBD is capped by an FD. The present results need to be confirmed in prospective studies.
Collapse
Affiliation(s)
| | - Eve Desmedt
- Service de Dermatologie, Hôpital C. Huriez, CHU de Lille
| | - Laure Deramoudt
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | - Michèle Vasseur
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | - Pascal Odou
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | | | - Bertrand Décaudin
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| | - Laurent Mortier
- Department of Dermatology, Claude Huriez Hospital, CARADERM and University of Lille, Lille, France
| | - Nicolas Simon
- Institut de Pharmacie, CHU Lille
- Univ. Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées
| |
Collapse
|
13
|
Lessard-Roy A, Marchand R, Lemieux P, Masse M, Lacerte A, Carmichael PH, Laurin D. Immune checkpoint inhibitors and risk of immune-mediated adverse events: a cohort study comparing extended versus standard interval administration. Clin Exp Med 2024; 24:40. [PMID: 38386053 PMCID: PMC10884063 DOI: 10.1007/s10238-024-01301-7] [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: 07/20/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
The COVID-19 pandemic precipitated the implementation of extended interval immune checkpoint inhibitors (ICIs) in an effort to limit hospital visits, but few studies have examined their safety. This study aimed to compare in oncology outpatients, immune-mediated adverse events (IMAEs) in terms of total number, incidence, severity, and time to occurrence, based on exposure to standard or extended interval ICIs. A retrospective cohort study was conducted in patients who received at least one dose of an ICI between 2015 and 2021. Data were collected from patient records and pharmacy software. Adjusted logistic, Poisson, and Cox regression models were estimated. A total of 310 patients with a mean age of 67.1 years were included, 130 of whom had the extended interval. No statistically significant differences were observed between the groups. With the standard and extended intervals, the mean total number of IMAE per participant was 1.02 and 1.18, respectively; the incidence of an IMAE was 62% and 64%. Of the 147 IMAE episodes in the standard interval group, 14 (9.5%) were grade 3 or higher, while there were 15 (12.4%) among the 121 IMAE episodes in the extended interval group. Compared with standard interval, the use of extended interval did not increase the risk of having a first IMAE (adjusted hazard ratio 0.92 (95% CI 0.67-1.26)). This study suggests that the administration of an ICI according to extended interval is as safe as the administration according to standard interval in oncology outpatients.
Collapse
Affiliation(s)
- Amélia Lessard-Roy
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Département de Pharmacie, Hôpital Sainte-Croix, Centre Intégré Universitaire de Santé et de Services Sociaux de la Mauricie-et-du-Centre-du-Québec, Drummondville, Canada
| | - Roxanne Marchand
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Département de Pharmacie, Centre Hospitalier Affilié Universitaire Régional de Trois-Rivières, Centre Intégré Universitaire de Santé et de Services Sociaux de la Mauricie-et-du-Centre-du-Québec, 1991 Boul. du Carmel, Trois-Rivières, QC, G8Z 3R9, Canada
| | - Pierre Lemieux
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
- Département de Pharmacie, Centre Hospitalier Affilié Universitaire Régional de Trois-Rivières, Centre Intégré Universitaire de Santé et de Services Sociaux de la Mauricie-et-du-Centre-du-Québec, 1991 Boul. du Carmel, Trois-Rivières, QC, G8Z 3R9, Canada.
| | - Mélanie Masse
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Département de Pharmacie, Centre Hospitalier Affilié Universitaire Régional de Trois-Rivières, Centre Intégré Universitaire de Santé et de Services Sociaux de la Mauricie-et-du-Centre-du-Québec, 1991 Boul. du Carmel, Trois-Rivières, QC, G8Z 3R9, Canada
| | | | - Pierre-Hugues Carmichael
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Centre d'excellence sur le Vieillissement de Québec, Centre Intégré Universitaire de Santé et de Services Sociaux de la Capitale Nationale, Québec, Canada
| | - Danielle Laurin
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
- Centre d'excellence sur le Vieillissement de Québec, Centre Intégré Universitaire de Santé et de Services Sociaux de la Capitale Nationale, Québec, Canada
| |
Collapse
|
14
|
Bai JPF, Stinchcomb AL, Wang J, Earp J, Stern S, Schuck RN. Creating a Roadmap to Quantitative Systems Pharmacology-Informed Rare Disease Drug Development: A Workshop Report. Clin Pharmacol Ther 2024; 115:201-205. [PMID: 37984065 DOI: 10.1002/cpt.3096] [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: 09/05/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
One of the goals of the Accelerating Rare Disease Cures (ARC) program in the Center for Drug Evaluation and Research (CDER) at the US Food and Drug Administration (FDA) is the development and use of regulatory and scientific tools, including drug/disease modeling, dose selection, and translational medicine tools. To facilitate achieving this goal, the FDA in collaboration with the University of Maryland Center of Excellence in Regulatory Science and Innovation (M-CERSI) hosted a virtual public workshop on May 11, 2023, entitled "Creating a Roadmap to Quantitative Systems Pharmacology-Informed Rare Disease Drug Development." This workshop engaged scientists from pharmaceutical companies, academic institutes, and the FDA to discuss the potential utility of quantitative systems pharmacology (QSP) in rare disease drug development and identify potential challenges and solutions to facilitate its use. Here, we report the main findings from this workshop, highlight the key takeaways, and propose a roadmap to facilitate the use of QSP in rare disease drug development.
Collapse
Affiliation(s)
- Jane P F Bai
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Jie Wang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Justin Earp
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Sydney Stern
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Robert N Schuck
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
15
|
Du S, Zhao Y, Hu Z, Liu S, Roy A, Shen J, Zhu L, Hamuro L. Pediatric model-based dose optimization using a pooled exposure-response safety analysis for nivolumab and nivolumab plus ipilimumab combination in melanoma. CPT Pharmacometrics Syst Pharmacol 2024; 13:168-179. [PMID: 37873561 PMCID: PMC10787196 DOI: 10.1002/psp4.13070] [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: 06/28/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023] Open
Abstract
An exposure-response (E-R) safety analysis was conducted across adult and pediatric (<18 years) studies to evaluate the potential impact of higher nivolumab and/or ipilimumab exposures in adolescents (≥12 to <18 years) versus adults with melanoma using the approved adult dosing regimens for nivolumab alone or in combination with ipilimumab. Data from 3507 patients across 15 studies were used to examine the relationship between nivolumab-ipilimumab daily average exposure and time to grade 2+ immune-mediated adverse events (gr2+ IMAEs). Results from the E-R safety model showed ipilimumab, but not nivolumab, exposure to be a statistically significant predictor of gr2+ IMAEs. Significant covariates included sex (41% higher risk for women than men), line of therapy (19% higher for first-line than later-line), and treatment setting (26% lower for adjuvant than advanced melanoma). Younger age and lower body weight (BW) were each associated with a lower risk of gr2+ IMAEs (hazard ratio [HR]: 0.830 for 15-year-olds versus 60-year-olds and 0.84 for BW 52 kg versus 75 kg). For adolescents with melanoma treated with nivolumab in the advanced or adjuvant settings, these results are supportive of nivolumab flat dosing regimens for adolescents greater than or equal to 40 kg and BW-based dosing for adolescents less than 40 kg. These results also support adult weight-based dosing regimens for nivolumab plus ipilimumab in adolescents with advanced melanoma. This analysis suggests that although higher exposures are predicted in adolescents with lower weight compared with adults, there is no predicted immune-mediated safety risk when treated with the approved adult dosing of nivolumab with/without ipilimumab.
Collapse
Affiliation(s)
| | - Yue Zhao
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Zheyi Hu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Sihang Liu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Amit Roy
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Jun Shen
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Li Zhu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | | |
Collapse
|
16
|
Ito T, Kaku-Ito Y, Ohno F, Nakahara T. A real-world study on the safety profile of extended-interval dosing of immune checkpoint inhibitors for melanoma: a single-center analysis in Japan. Front Med (Lausanne) 2023; 10:1293397. [PMID: 38143437 PMCID: PMC10740208 DOI: 10.3389/fmed.2023.1293397] [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: 09/13/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Background Anti-programmed death-1 (PD-1) antibodies are the mainstay for the treatment of unresectable or high-risk melanoma. However, real-world data on the safety profile of their extended-interval doses (EDs) are limited, particularly in Asian patients with melanoma. Materials and methods In this single-center retrospective study, we analyzed the risks of immune-related adverse events (irAEs) among 71 Japanese patients (36 males; mean age, 65.0 years) who received anti-PD-1 monotherapy for melanoma at our institute. Patients who were administered ipilimumab prior to anti-PD-1 monotherapy were excluded. Patients were divided into three groups: canonical-interval dose (CD) group (n = 50, body weight-based dosing or 240 mg Q2W for nivolumab and body weight-based dosing or 200 mg Q3W for pembrolizumab), ED group (n = 14, 480 mg Q4W for nivolumab and 400 mg Q6W for pembrolizumab), and dose-switch (DS) group (n = 7, upfront CD followed by ED). Results The CD group received nivolumab more frequently in the metastatic setting. There were no significant differences in baseline characteristics among the three groups, including in sex, age, primary tumor site, tumor subtype, and follow-up period. irAEs occurred in 36.6% (26 patients) of all patients (32.0% of the CD group, 35.7% of the ED group, and 71.4% of the DS group), while severe (grade ≥ 3) irAEs occurred in only two patients, both of whom were in the CD group. Most of the irAEs occurred during the first 6 months of anti-PD-1 therapy and, interestingly, all of the irAEs in the DS group occurred before the switch (during the CD). There was no significant difference among the three groups in the probability of irAE estimated by the Kaplan-Meier method. Conclusion These findings may highlight the safety of ED of anti-PD-1 monotherapy in the treatment of Asian patients with melanoma.
Collapse
Affiliation(s)
- Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | |
Collapse
|
17
|
Parvez A, Choudhary F, Mudgal P, Khan R, Qureshi KA, Farooqi H, Aspatwar A. PD-1 and PD-L1: architects of immune symphony and immunotherapy breakthroughs in cancer treatment. Front Immunol 2023; 14:1296341. [PMID: 38106415 PMCID: PMC10722272 DOI: 10.3389/fimmu.2023.1296341] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/08/2023] [Indexed: 12/19/2023] Open
Abstract
PD-1 (Programmed Cell Death Protein-1) and PD-L1 (Programmed Cell Death Ligand-1) play a crucial role in regulating the immune system and preventing autoimmunity. Cancer cells can manipulate this system, allowing them to escape immune detection and promote tumor growth. Therapies targeting the PD-1/PD-L1 pathway have transformed cancer treatment and have demonstrated significant effectiveness against various cancer types. This study delves into the structure and signaling dynamics of PD-1 and its ligands PD-L1/PD-L2, the diverse PD-1/PD-L1 inhibitors and their efficacy, and the resistance observed in some patients. Furthermore, this study explored the challenges associated with the PD-1/PD-L1 inhibitor treatment approach. Recent advancements in the combination of immunotherapy with chemotherapy, radiation, and surgical procedures to enhance patient outcomes have also been highlighted. Overall, this study offers an in-depth overview of the significance of PD-1/PD-L1 in cancer immunotherapy and its future implications in oncology.
Collapse
Affiliation(s)
- Adil Parvez
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia, Hamdard, New Delhi, India
| | - Furqan Choudhary
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia, Hamdard, New Delhi, India
| | - Priyal Mudgal
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia, Hamdard, New Delhi, India
| | - Rahila Khan
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia, Hamdard, New Delhi, India
| | - Kamal A. Qureshi
- Department of Pharmaceutics, Unaizah College of Pharmacy, Qassim University, Unaizah, Qassim, Saudi Arabia
| | - Humaira Farooqi
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia, Hamdard, New Delhi, India
| | - Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
18
|
Colard-Thomas J, Manceron C, Duflos C, Herman F, Simon M, Maria ATJ, Faillie JL, Viala M, Palassin P. Comparison of clinical safety between standard versus extended interval dosing of immune checkpoint inhibitors: a real-world retrospective cohort study. ESMO Open 2023; 8:102070. [PMID: 37988951 PMCID: PMC10774961 DOI: 10.1016/j.esmoop.2023.102070] [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: 06/13/2023] [Revised: 10/15/2023] [Accepted: 10/21/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Extended interval dosing (ED) for inhibitors of programmed cell death protein 1 (anti-PD-1) (nivolumab, pembrolizumab) or its ligand (anti-PD-L1) (durvalumab) were recently approved based on pharmacokinetic model results that predicted a benefit-risk profile comparable with the standard dosing (SD) regimen. However, safety data in real-world condition of use are lacking. The objective was to compare the incidence and the risk factors of serious immune-related adverse events (irAEs) and any-grade irAEs between the SD and ED regimens in patients treated with anti-PD-1 or anti-PD-L1. MATERIALS AND METHODS IrAEs were assessed from medical records in all new users of nivolumab, pembrolizumab, or durvalumab between 1 January 2019 and 31 December 2020 across two oncology centers in France. The incidence of irAEs was compared between both dosing regimens using Cox proportional hazards models adjusting for the main available confounders. RESULTS Among 686 patients included, 63% were new users of an SD regimen, 14% of ED regimen, and 23% started with SD and switched to ED regimen during follow-up. Overall, 34.6% of patients experienced at least one irAE of any grade and 11.4% presented at least one serious grade ≥3 irAE. No statistical difference was found between the SD and ED regimen on the risk of grade ≥3 irAEs [adjusted hazard ratio (HR) 1.40, 95% confidence interval (CI) 0.71-2.76] but our results suggest an increased risk of any-grade irAEs with the ED regimen (adjusted HR 1.46, 95% CI 1.00-2.12, P = 0.048). IrAEs resolved without sequelae in 46.4% of cases, and they were fatal for three patients (0.4%). Autoimmune pre-existing condition was confirmed as a risk factor for grade ≥3 irAEs (HR 2.56, 95% CI 1.53-4.27) and for all-grade irAEs (HR 1.60, 95% CI 1.17-2.20). CONCLUSIONS In a real-world setting, according to the regimen chosen by the oncologist based on clinical characteristics, we did not observe an increase in grade ≥3 irAE incidence between the SD and ED regimens.
Collapse
Affiliation(s)
- J Colard-Thomas
- Department of Medical Oncology, Montpellier Cancer Institute, Univ Montpellier, Montpellier, France.
| | - C Manceron
- Department of Medical Pharmacology and Toxicology, CHU Montpellier, University of Montpellier, Montpellier, France
| | - C Duflos
- Department of Medical Information, Public Health Unit, CHU Montpellier, Univ Montpellier, Montpellier, France
| | - F Herman
- Department of Medical Information, Public Health Unit, CHU Montpellier, Univ Montpellier, Montpellier, France
| | - M Simon
- Department of UPCO, CHU Montpellier, Univ Montpellier, Montpellier, France
| | - A T J Maria
- Internal Medicine & Immuno-Oncology (MedI(2)O), Institute for Regenerative Medicine and Biotherapy (IRMB), Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France; University of Montpellier, IRMB, Inserm U1183, Montpellier, France
| | - J-L Faillie
- Department of Medical Pharmacology and Toxicology, CHU Montpellier, University of Montpellier, Montpellier, France; University of Montpellier, IDESP INSERM, Montpellier, France
| | - M Viala
- Department of Medical Oncology, Montpellier Cancer Institute, Univ Montpellier, Montpellier, France
| | - P Palassin
- Department of Medical Pharmacology and Toxicology, CHU Montpellier, University of Montpellier, Montpellier, France
| |
Collapse
|
19
|
Marolleau S, Mogenet A, Boeri C, Hamimed M, Ciccolini J, Greillier L. Killing a fly with a sledgehammer: Atezolizumab exposure in real-world lung cancer patients. CPT Pharmacometrics Syst Pharmacol 2023; 12:1795-1803. [PMID: 38011601 PMCID: PMC10681534 DOI: 10.1002/psp4.13063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/25/2023] [Accepted: 10/02/2023] [Indexed: 11/29/2023] Open
Abstract
Atezolizumab is an anti-PDL1 approved for treating lung cancer. A threshold of 6 μg/mL in plasma has been associated with target engagement. The extent to which patients could be overexposed with the standard 1200 mg q3w dosing remains unknown. Here, we monitored atezolizumab peak and trough levels in 27 real-world patients with lung cancer as part of routine therapeutic drug monitoring. Individual pharmacokinetic (PK) parameters were calculated using a population approach and optimal dosing-intervals were simulated with respect to the target trough levels. No patient had plasma levels below 6 μg/mL. The results showed that the mean trough level after the first treatment was 78.3 ± 17 μg/mL, that is, 13 times above the target concentration. The overall response rate was 55.5%. Low-grade immune-related adverse events was observed in 37% of patients. No relationship was found between exposure metrics of atezolizumab (i.e., minimum plasma concentration, maximum plasma concentration, and area under the curve) and pharmacodynamic end points (i.e., efficacy and toxicity). Further simulations suggest that the dosing interval could be extended from 21 days to 49 up to 136 days (mean: 85.7 days, i.e., q12w), while ensuring plasma levels still above the 6 μg/mL target threshold. This observational, real-world study suggests that the standard 1200 mg q3w fixed-dose regimen of atezolizumab results in significant overexposure in all the patients. This was not associated with increased side effects. As plasma levels largely exceed pharmacologically active concentrations, interindividual variability in PK parameters did not impact efficacy. Our data suggest that dosing intervals could be markedly extended with respect to the target threshold associated with efficacy.
Collapse
Affiliation(s)
- Sophie Marolleau
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Alice Mogenet
- Oncologie multidisciplinaire et innovations thérapeutiquesNord University Hospital of MarseilleMarseilleFrance
| | - Clara Boeri
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Mourad Hamimed
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Joseph Ciccolini
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Laurent Greillier
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
- Oncologie multidisciplinaire et innovations thérapeutiquesNord University Hospital of MarseilleMarseilleFrance
| |
Collapse
|
20
|
Becker JC, Ugurel S, Leiter U, Meier F, Gutzmer R, Haferkamp S, Zimmer L, Livingstone E, Eigentler TK, Hauschild A, Kiecker F, Hassel JC, Mohr P, Fluck M, Thomas I, Garzarolli M, Grimmelmann I, Drexler K, Spillner AN, Eckhardt S, Schadendorf D. Adjuvant immunotherapy with nivolumab versus observation in completely resected Merkel cell carcinoma (ADMEC-O): disease-free survival results from a randomised, open-label, phase 2 trial. Lancet 2023; 402:798-808. [PMID: 37451295 DOI: 10.1016/s0140-6736(23)00769-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Merkel cell carcinoma (MCC) is an immunogenic but aggressive skin cancer. Even after complete resection and radiation, relapse rates are high. PD-1 and PD-L1 checkpoint inhibitors showed clinical benefit in advanced MCC. We aimed to assess efficacy and safety of adjuvant immune checkpoint inhibition in completely resected MCC (ie, a setting without an established systemic standard-of-care treatment). METHODS In this multicentre phase 2 trial, patients (any stage, Eastern Cooperative Oncology Group performance status 0-1) at 20 academic medical centres in Germany and the Netherlands with completely resected MCC lesions were randomly assigned 2:1 to receive nivolumab 480 mg every 4 weeks for 1 year, or observation, stratified by stage (American Joint Committee on Cancer stages 1-2 vs stages 3-4), age (<65 vs ≥65 years), and sex. Landmark disease-free survival (DFS) at 12 and 24 months was the primary endpoint, assessed in the intention-to-treat populations. Overall survival and safety were secondary endpoints. This planned interim analysis was triggered when the last-patient-in was followed up for more than 1 year. This study is registered with ClinicalTrials.gov (NCT02196961) and with the EU Clinical Trials Register (2013-000043-78). FINDINGS Between Oct 1, 2014, and Aug 31, 2020, 179 patients were enrolled (116 [65%] stage 3-4, 122 [68%] ≥65 years, 111 [62%] male). Stratification factors (stage, age, sex) were balanced across the nivolumab (n=118) and internal control group (observation, n=61); adjuvant radiotherapy was more common in the control group. At a median follow-up of 24·3 months (IQR 19·2-33·4), median DFS was not reached (between-groups hazard ratio 0·58, 95% CI 0·30-1·12); DFS rates in the nivolumab group were 85% at 12 months and 84% at 24 months, and in the observation group were 77% at 12 months and 73% at 24 months. Overall survival results were not yet mature. Grade 3-4 adverse events occurred in 48 [42%] of 115 patients who received at least one dose of nivolumab and seven [11%] of 61 patients in the observation group. No treatment-related deaths were reported. INTERPRETATION Adjuvant therapy with nivolumab resulted in an absolute risk reduction of 9% (1-year DFS) and 10% (2-year DFS). The present interim analysis of ADMEC-O might suggest clinical use of nivolumab in this area of unmet medical need. However, overall survival events rates, with ten events in the active treatment group and six events in the half-the-size observation group, are not mature enough to draw conclusions. The explorative data of our trial support the continuation of ongoing, randomised trials in this area. ADMEC-O suggests that adjuvant immunotherapy is clinically feasible in this area of unmet medical need. FUNDING Bristol Myers Squibb.
Collapse
Affiliation(s)
- Jürgen C Becker
- Department of Dermatology, University Hospital Essen, Essen, Germany; Translational Skin Cancer Research, Department of Dermatology and West German Cancer Center, University of Medicine Duisburg-Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Ulrike Leiter
- Centre for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany; German Cancer Consortium, Partner Site Tübingen, Tübingen, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Dresden, Dresden, Germany; German Cancer Consortium, Partner Site Dresden, Dresden, Germany
| | - Ralf Gutzmer
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum, Minden, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany
| | - Thomas K Eigentler
- Centre for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany; Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Felix Kiecker
- Department of Dermatology and Venereology, Vivantes Klinikum Berlin Neukölln, Berlin, Germany; Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jessica C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium, Partner Site Heidelberg, Heidelberg, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Kliniken, Buxtehude, Germany
| | - Michael Fluck
- Department of Oncology Hornheide, Fachklinik Hornheide, Münster, Germany
| | - Ioannis Thomas
- Centre for Dermatooncology, Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Marlene Garzarolli
- Department of Dermatology, University Hospital Dresden, Dresden, Germany
| | - Imke Grimmelmann
- Skin Cancer Center Hannover, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Konstantin Drexler
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | | | | | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Partner Site Essen, Essen, Germany.
| |
Collapse
|
21
|
Barrett JS, Goyal RK, Gobburu J, Baran S, Varshney J. An AI Approach to Generating MIDD Assets Across the Drug Development Continuum. AAPS J 2023; 25:70. [PMID: 37430126 DOI: 10.1208/s12248-023-00838-x] [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: 03/17/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023] Open
Abstract
Model-informed drug development involves developing and applying exposure-based, biological, and statistical models derived from preclinical and clinical data sources to inform drug development and decision-making. Discrete models are generated from individual experiments resulting in a single model expression that is utilized to inform a single stage-gate decision. Other model types provide a more holistic view of disease biology and potentially disease progression depending on the appropriateness of the underlying data sources for that purpose. Despite this awareness, most data integration and model development approaches are still reliant on internal (within company) data stores and traditional structural model types. An AI/ML-based MIDD approach relies on more diverse data and is informed by past successes and failures including data outside a host company (external data sources) that may enhance predictive value and enhance data generated by the sponsor to reflect more informed and timely experimentation. The AI/ML methodology also provides a complementary approach to more traditional modeling efforts that support MIDD and thus yields greater fidelity in decision-making. Early pilot studies support this assessment but will require broader adoption and regulatory support for more evidence and refinement of this paradigm. An AI/ML-based approach to MIDD has the potential to transform regulatory science and the current drug development paradigm, optimize information value, and increase candidate and eventually product confidence with respect to safety and efficacy. We highlight early experiences with this approach using the AI compute platforms as representative examples of how MIDD can be facilitated with an AI/ML approach.
Collapse
Affiliation(s)
- Jeffrey S Barrett
- Aridhia Bioinformatics, 163 Bath Street, Glasgow, Scotland, G2 4SQ, UK.
| | - Rahul K Goyal
- Center for Translational Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Jogarao Gobburu
- Center for Translational Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA
- Pumas-AI, Baltimore, Maryland, USA
| | | | | |
Collapse
|
22
|
Cantini L, Paoloni F, Pecci F, Spagnolo F, Genova C, Tanda ET, Aerts S, Rebuzzi SE, Fornarini G, Zoratto F, Fancelli S, Lupi A, Della Corte CM, Parisi A, Bennati C, Ortega C, Atzori F, Piovano PL, Orciuolo C, De Tursi M, Ghidini M, Botticelli A, Scagnoli S, Belluomini L, Leporati R, Veccia A, Di Giacomo AM, Festino L, Cortinovis D, Acquati M, Filetti M, Giusti R, Tucci M, Sergi MC, Garutti M, Puglisi F, Manglaviti S, Citarella F, Santoni M, Rijavec E, Lo Russo G, Santini D, Addeo A, Antonuzzo L, Indini A, Rocchi MBL, Cortellini A, Grossi F, Ascierto PA, Aerts JGJV, Berardi R. Safety of extended interval dosing immune checkpoint inhibitors: a multicenter cohort study. J Natl Cancer Inst 2023; 115:796-804. [PMID: 37042716 PMCID: PMC10323889 DOI: 10.1093/jnci/djad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/10/2023] [Accepted: 03/24/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Real-life spectrum and survival implications of immune-related adverse events (irAEs) in patients treated with extended interval dosing (ED) immune checkpoint inhibitors (ICIs) are unknown. METHODS Characteristics of 812 consecutive solid cancer patients who received at least 1 cycle of ED monotherapy (pembrolizumab 400 mg Q6W or nivolumab 480 mg Q4W) after switching from canonical interval dosing (CD; pembrolizumab 200 mg Q3W or nivolumab 240 mg Q2W) or treated upfront with ED were retrieved. The primary objective was to compare irAEs patterns within the same population (before and after switch to ED). irAEs spectrum in patients treated upfront with ED and association between irAEs and overall survival were also described. RESULTS A total of 550 (68%) patients started ICIs with CD and switched to ED. During CD, 225 (41%) patients developed any grade and 17 (3%) G3 or G4 irAEs; after switching to ED, any grade and G3 or G4 irAEs were experienced by 155 (36%) and 20 (5%) patients. Switching to ED was associated with a lower probability of any grade irAEs (adjusted odds ratio [aOR] = 0.83, 95% confidence interval [CI] = 0.64 to 0.99; P = .047), whereas no difference for G3 or G4 events was noted (aOR = 1.55, 95% CI = 0.81 to 2.94; P = .18). Among patients who started upfront with ED (n = 232, 32%), 107 (41%) developed any grade and 14 (5%) G3 or G4 irAEs during ED. Patients with irAEs during ED had improved overall survival (adjusted hazard ratio [aHR] = 0.53, 95% CI = 0.34 to 0.82; P = .004 after switching; aHR = 0.57, 95% CI = 0.35 to 0.93; P = .025 upfront). CONCLUSIONS Switching ICI treatment from CD and ED did not increase the incidence of irAEs and represents a safe option also outside clinical trials.
Collapse
Affiliation(s)
- Luca Cantini
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
- Labcorp Drug Development Inc, Princeton, NJ, USA
| | - Francesco Paoloni
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - Federica Pecci
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - Francesco Spagnolo
- Medical Oncology Unit 2, Istituto di ricovero e cura a carattere scientific (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Carlo Genova
- Academic Medical Oncology Unit, Istituto di ricovero e cura a carattere scientific (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genoa, Italy
| | - Enrica Teresa Tanda
- Medical Oncology Unit 2, Istituto di ricovero e cura a carattere scientific (IRCCS) Ospedale Policlinico San Martino, Genova, Italy
| | - Sophie Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Sara Elena Rebuzzi
- Department of Internal Medicine and Medical Specialties, School of Medicine, University of Genoa, Italy
- Medical Oncology Unit, Ospedale San Paolo, Savona, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, Istituto di ricovero e cura a carattere scientific (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Sara Fancelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Alessio Lupi
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | | | - Alessandro Parisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Chiara Bennati
- S Maria delle Croci Hospital, AUSL della Romagna, Ravenna, Italy
| | - Cinzia Ortega
- Oncology, Asl Cn2, Ospedale Michele e Pietro Ferrero, Verduno, Italy
| | - Francesco Atzori
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari, Italy
| | - Pier Luigi Piovano
- Oncology Unit, Azienda Ospedaliera “SS. Antonio e Biagio e C. Arrigo”, Alessandria, Italy
| | - Corrado Orciuolo
- Istituto di ricovero e cura a carattere scientific (IRCCS), National Cancer Institute Regina Elena, Rome, Italy
| | - Michele De Tursi
- Department of Innovative Technologies in Medicine and Dentistry, University G. D’Annunzio, Chieti-Pescara, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione Istituto di ricovero e cura a carattere scientific (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Andrea Botticelli
- Medical Oncology Unit A, Policlinico Umberto I, Radiological, Oncological, Pathological Sciences Department, Sapienza University of Rome, Italy
| | | | - Lorenzo Belluomini
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Rita Leporati
- Medical Oncology, Fondazione Istituto di ricovero e cura a carattere scientific (IRCCS) Istituto Nazionale dei Tumori, Milano, Italy
| | - Antonello Veccia
- Medical Oncology, Santa Chiara Hospital, Largo Medaglie d’Oro 1, Trento, Italy
| | - Anna Maria Di Giacomo
- Center for Immuno-Oncology, University of Siena, University Hospital of Siena, Siena, Italy
| | - Lucia Festino
- Melanoma Unit, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori Istituto di ricovero e cura a carattere scientific (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | | | | | - Marco Filetti
- Phase 1 Unit, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di ricovero e cura a carattere scientific (IRCCS), Rome, Italy
| | | | - Marco Tucci
- Medical Oncology Unit, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Italy
| | - Maria Chiara Sergi
- Medical Oncology Unit, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Italy
| | - Mattia Garutti
- CRO Aviano, National Cancer Institute, Istituto di ricovero e cura a carattere scientific (IRCCS), Aviano, Italy
| | - Fabio Puglisi
- CRO Aviano, National Cancer Institute, Istituto di ricovero e cura a carattere scientific (IRCCS), Aviano, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Sara Manglaviti
- Thoracic Unit, Medical Oncology Department 1, Fondazione Istituto di ricovero e cura a carattere scientific (IRCCS) Istituto Nazionale dei Tumori di Milano, Italy
| | - Fabrizio Citarella
- Department of Medical Oncology, Campus Bio-Medico University, Rome, Italy
| | | | - Erika Rijavec
- Medical Oncology Unit, Ospedale di Circolo e Fondazione Macchi, asst Settelaghi, Varese, Italy
| | - Giuseppe Lo Russo
- Thoracic Unit, Medical Oncology Department 1, Fondazione Istituto di ricovero e cura a carattere scientific (IRCCS) Istituto Nazionale dei Tumori di Milano, Italy
| | - Daniele Santini
- UOC Oncologia Medica Territoriale, Sapienza Università, Polo Pontino, Rome, Italy
| | - Alfredo Addeo
- Oncology Department, University Hospital Geneva, Geneva, Switzerland
| | - Lorenzo Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Clinical Oncology Unit, and Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Alice Indini
- Medical Oncology Unit, Ospedale di Circolo e Fondazione Macchi, asst Settelaghi, Varese, Italy
| | | | - Alessio Cortellini
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Francesco Grossi
- Medical Oncology Unit, Ospedale di Circolo e Fondazione Macchi, asst Settelaghi, University of insubria, Varese, Italy
| | - Paolo Antonio Ascierto
- Melanoma Unit, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori Istituto di ricovero e cura a carattere scientific (IRCCS) Fondazione “G. Pascale”, Naples, Italy
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Rossana Berardi
- Clinical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| |
Collapse
|
23
|
Javid H, Attarian F, Saadatmand T, Rezagholinejad N, Mehri A, Amiri H, Karimi-Shahri M. The therapeutic potential of immunotherapy in the treatment of breast cancer: Rational strategies and recent progress. J Cell Biochem 2023; 124:477-494. [PMID: 36966454 DOI: 10.1002/jcb.30402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/25/2023] [Accepted: 03/12/2023] [Indexed: 03/27/2023]
Abstract
The second leading cause of cancer death in women worldwide is breast cancer (BC), and despite significant advances in BC therapies, a significant proportion of patients develop metastasis and disease recurrence. Currently used treatments, like radiotherapy, chemotherapy, and hormone replacement therapy, result in poor responses and high recurrence rates. Alternative therapies are therefore needed for this type of cancer. Cancer patients may benefit from immunotherapy, a novel treatment strategy in cancer treatment. Even though immunotherapy has been successful in many cases, some patients do not respond to the treatment or those who do respond relapse or progress. The purpose of this review is to discuss several different immunotherapy approaches approved for the treatment of BC, as well as different strategies for immunotherapy for the treatment of BC.
Collapse
Affiliation(s)
- Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Attarian
- Department of Biology, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Toktam Saadatmand
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | | | - Ali Mehri
- Endoscopic and Minimally Invasive Surgery Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Amiri
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
24
|
A comprehensive regulatory and industry review of modeling and simulation practices in oncology clinical drug development. J Pharmacokinet Pharmacodyn 2023; 50:147-172. [PMID: 36870005 PMCID: PMC10169901 DOI: 10.1007/s10928-023-09850-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 02/16/2023] [Indexed: 03/05/2023]
Abstract
Exposure-response (E-R) analyses are an integral component in the development of oncology products. Characterizing the relationship between drug exposure metrics and response allows the sponsor to use modeling and simulation to address both internal and external drug development questions (e.g., optimal dose, frequency of administration, dose adjustments for special populations). This white paper is the output of an industry-government collaboration among scientists with broad experience in E-R modeling as part of regulatory submissions. The goal of this white paper is to provide guidance on what the preferred methods for E-R analysis in oncology clinical drug development are and what metrics of exposure should be considered.
Collapse
|
25
|
Schlüter J, Cunningham S, Zimmermann R, Achenbach S, Kramer R, Erdmann M, Beckmann M, Heinzerling L, Hackstein H. Characterization of the impact of immune checkpoint inhibitors on platelet activation and aggregation. Immunobiology 2023; 228:152311. [PMID: 36495598 DOI: 10.1016/j.imbio.2022.152311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are effective oncological drugs which block cellular check-point receptors typically targeted by tumor immune evasion strategies. Despite their benefits, clinicians have reported treatment-associated thromboembolism during ICI therapy in recent years. Though several theories on this ICI-associated pathogenesis exist, the direct effects of ICIs on platelets remains unknown. We therefore investigated the potential direct and indirect effect of PD-1, PD-L1 and CTLA-4-targeting ICIs on platelet functionality in multifaceted in vitro experiments. Interestingly, we could not observe a clear effect of ICI on platelet aggregation and primary hemostasis in whole blood and platelet concentrate-based assays. Furthermore, the presence of ICIs in toll-like receptor stimulation had no significant impact on platelet surface marker expression. In a second approach, we investigated the indirect immunological impact of ICIs on platelet activation by exposing platelets to supernatants from ICI- and Staphylococcal enterotoxin B-exposed PBMCs. Whereas ICIs affected IL-2 levels in supernatants, we could not detect clear differences in the secretion of pro-thrombogenic factors and platelet responses. The obtained data suggest that the direct influence of ICIs on platelet activation or the influence of altered T cell function on platelet activation cannot be considered a major factor in the development of thrombotic events.
Collapse
Affiliation(s)
- Julian Schlüter
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany.
| | - Robert Zimmermann
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| | - Susanne Achenbach
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| | - Rafaela Kramer
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany
| | - Michael Erdmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany
| | - Malte Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany
| | - Lucie Heinzerling
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany; Department of Dermatology and Allergology, Ludwig-Maximilian University, Munich 80539, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| |
Collapse
|
26
|
McQuade JL, Hammers H, Furberg H, Engert A, André T, Blumenschein G, Tannir N, Baron A, Larkin J, El-Khoueiry A, Carbone DP, Thomas JM, Hennicken D, Coffey M, Motzer RJ. Association of Body Mass Index With the Safety Profile of Nivolumab With or Without Ipilimumab. JAMA Oncol 2023; 9:102-111. [PMID: 36480191 PMCID: PMC9857666 DOI: 10.1001/jamaoncol.2022.5409] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Increased survival with immune checkpoint inhibitors has been reported for patients with obesity vs a normal body mass index (BMI). However, the association of obesity with the safety of immune checkpoint inhibitors warrants study. Objective To investigate associations between BMI and immune-related adverse events (irAEs) among patients with advanced cancers treated with nivolumab monotherapy and nivolumab plus ipilimumab combination therapy. Design, Setting, and Participants This study was a retrospective pooled analysis of 3772 patients from 14 multicenter CheckMate clinical trials across 8 tumor types. Patients with advanced cancers received nivolumab, 3 mg/kg (n = 2746); nivolumab, 3 mg/kg, plus ipilimumab, 1 mg/kg (n = 713); or nivolumab, 1 mg/kg, plus ipilimumab, 3 mg/kg (n = 313). Baseline BMI was categorized as normal weight or underweight (<25), overweight (25 to <30), or obese (≥30) according to World Health Organization criteria. The studies began patient enrollment between February 9, 2012, and May 21, 2015, and patients were followed up to database lock on May 1, 2019. Data analysis was conducted from May 1 to September 1, 2019. Interventions Nivolumab, 3 mg/kg; nivolumab, 3 mg/kg, plus ipilimumab, 1 mg/kg; and nivolumab, 1 mg/kg, plus ipilimumab, 3 mg/kg. Main Outcomes and Measures Odds ratios (ORs) and 95% CIs for incidence of any-grade and grade 3 or 4 irAEs were calculated for patients with obesity vs normal weight or underweight BMI in the overall cohort and in subgroups based on patient and tumor characteristics. Analyses for nivolumab plus ipilimumab cohorts were exploratory. Results A total of 3772 patients were included, 2600 were male (69%), and median age was 61 years (range, 18-90 years). For patients receiving monotherapy with nivolumab, 3 mg/kg (n = 2746), the incidence of any-grade irAEs was higher in patients with obesity (n = 543) vs those with normal weight or underweight BMI (n = 1266; OR, 1.71; 95% CI, 1.38-2.11). Incidence of grade 3 or 4 irAEs did not differ between patients with obesity and those with normal weight or underweight BMI (OR, 1.21; 95% CI, 0.92-1.61). Risk of any-grade and grade 3 or 4 irAEs appeared consistent with that in the overall population across all subgroups evaluated except for a higher likelihood of grade 3 or 4 irAEs among female patients with obesity vs normal weight or underweight BMI (OR, 1.73; 95% CI, 1.07-2.79). For patients receiving nivolumab plus ipilimumab, the incidence of irAEs appeared consistent across BMI categories. Conclusions and Relevance Obesity appeared to be associated with an increased incidence of any-grade irAEs among patients treated with nivolumab monotherapy and with grade 3 or 4 irAEs among female patients only. These findings may inform the monitoring of patients at high risk of developing irAEs.
Collapse
Affiliation(s)
- Jennifer L McQuade
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Hans Hammers
- Kidney Cancer Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas
| | - Helena Furberg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andreas Engert
- German Hodgkin Study Group (GHSG), University Hospital of Cologne, Cologne, Germany
| | - Thierry André
- Department of Medical Oncology, Sorbonne University and Saint-Antoine Hospital, Paris, France
| | - George Blumenschein
- Department of Thoracic Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Nizar Tannir
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Ari Baron
- Division of Hematology Oncology, California Pacific Medical Center, San Francisco
| | - James Larkin
- Department of Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Anthony El-Khoueiry
- Department of Medicine, USC Norris Comprehensive Cancer Center, Los Angeles, California
| | - David P Carbone
- Department of Internal Medicine, Division of Medical Oncology, Ohio State University, Columbus
| | | | | | | | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
27
|
Malmberg R, Zietse M, Dumoulin DW, Hendrikx JJMA, Aerts JGJV, van der Veldt AAM, Koch BCP, Sleijfer S, van Leeuwen RWF. Alternative dosing strategies for immune checkpoint inhibitors to improve cost-effectiveness: a special focus on nivolumab and pembrolizumab. Lancet Oncol 2022; 23:e552-e561. [DOI: 10.1016/s1470-2045(22)00554-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022]
|
28
|
Lu S, Cheng Y, Zhou J, Wang M, Zhao J, Wang B, Chen G, Feng J, Ma Z, Wu L, Wang C, Ma K, Zhang S, Liang J, Song Y, Wang J, Wu YL, Li A, Huang Y, Chang J. An open label, safety study of Asian patients with advanced non-small-cell lung cancer receiving second-line nivolumab monotherapy (CheckMate 870). Ther Adv Med Oncol 2022; 14:17588359221138380. [PMID: 36425873 PMCID: PMC9679349 DOI: 10.1177/17588359221138380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/25/2022] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND Nivolumab has been approved in China as second-line treatment for advanced non-small-cell lung cancer (NSCLC) via weight-based infusion, based on the CheckMate 078 study. We investigated the safety and efficacy of 240 mg flat-dose nivolumab in patients with advanced NSCLC, including those with hepatitis B virus (HBV) and epidermal growth factor receptor (EGFR) mutation/ALK receptor tyrosine kinase (ALK) translocation due to high prevalence in China. METHODS CheckMate 870 was a single-arm, open-label, phase IIIb trial in Asian (primarily Chinese) patients with previously treated advanced NSCLC. Patients received flat-dose nivolumab 240 mg every 2 weeks (Q2W) for up to 2 years. The primary endpoint was the incidence and severity of treatment-related select adverse events (TRsAEs) in non-HBV patients; secondary and exploratory endpoints included severity of high-grade TRsAEs in HBV-infected patients, and safety, efficacy and patient-reported outcomes (PROs) in the whole population. RESULTS Out of 404 patients enrolled, 400 received treatment. Median (standard deviation) age was 60.5 (8.68) years and the majority were male (78.5%). At a median follow-up of 37.6 months, no Grade 5 TRsAEs were reported, and the frequency of Grade 3-4 TRsAEs was low (0.0-5.9%) in non-HBV and HBV NSCLC patients. Median overall survival (OS) and progression-free survival (PFS) in all treated patients were 14.7 (12.3-18.1) and 3.6 (2.3-3.8) months, respectively. Median OS was 14.2 (12.3-18.1) and 22.3 (10.0-NA) months for non-HBV and HBV-infected patients, 19.3 (11.2-31.7) and 13.7 (11.5-18.1) months for EGFR-positive and wild-type subgroups, and 19.3 (12.9-23.5) and 13.3 (10.9-17.7) months for those with programmed death-ligand 1 (PD-L1) expression ⩾1% and <1%, respectively. No notable changes from baseline were observed in PROs throughout the study. CONCLUSION Nivolumab 240 mg infusion Q2W was well tolerated, efficacious, and maintained health status and quality of life in Asian patients with previously treated advanced NSCLC regardless of HBV, EGFR, or PD-L1 status.
Collapse
Affiliation(s)
- Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, 241 Huaihai West Road, Xuhui District, Shanghai, China
| | | | - Jianying Zhou
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Mengzhao Wang
- Peking Union Medical College Hospital, Beijing, China
| | - Jun Zhao
- Beijing Cancer Hospital, Beijing, China
| | | | - Gongyan Chen
- Affiliated Cancer Hospital of Harbin Medical University, Harbin, China
| | - Jifeng Feng
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Zhiyong Ma
- Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Lin Wu
- Hunan Cancer Hospital (The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University), Changsha, China
| | - Changli Wang
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Kewei Ma
- The First Hospital of Jilin University, Changchun, China
| | | | - Jun Liang
- Peking University International Hospital, Beijing, China
| | - Yong Song
- Nanjing General Hospital, Nanjing, China
| | - Jie Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ang Li
- Bristol Myers Squibb Company, Princeton, NJ, USA
| | | | - Jianhua Chang
- Shanghai Cancer Center Fudan University, Shanghai, China
| |
Collapse
|
29
|
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
|
30
|
Samlowski W, Robert NJ, Chen L, Schenkel B, Davis C, Moshyk A, Kotapati S, Poretta T, Weber JS. Real-World nivolumab dosing patterns and safety outcomes in patients receiving adjuvant therapy for melanoma. Cancer Med 2022; 12:2378-2388. [PMID: 35880244 PMCID: PMC9939122 DOI: 10.1002/cam4.5061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/10/2022] [Accepted: 07/08/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Nivolumab at a dose of 480 mg every 4 weeks (Q4W) is approved for the adjuvant treatment of melanoma. However, real-world data on this regimen are limited in this setting. METHODS This retrospective observational study utilized data from the US Oncology Network iKnowMed electronic health record database and patient medical charts. Eligible patients were diagnosed with melanoma and received adjuvant nivolumab monotherapy from March to August 2018. Patients were grouped by dosing regimen: cohort 1 (C1), de novo nivolumab 480 mg Q4W; cohort 2 (C2), switched to nivolumab 480 mg Q4W after nivolumab 240 mg or 3 mg/kg every 2 weeks (Q2W); cohort 3 (C3), de novo nivolumab 3 mg/kg Q2W; or cohort 4 (C4), de novo nivolumab 240 mg Q2W. Patients were followed for up to 12 months. Duration of therapy and safety/tolerability were assessed. RESULTS One hundred ninety-one patients were included (C1, n = 40; C2, n = 74; C3, n = 22; C4, n = 55). Duration of therapy was relatively consistent across cohorts (median, 10.3 months; range, 8.3-10.7). Likewise, proportions of patients experiencing treatment-related adverse events (TRAEs) were similar (range, 54.5%-60.1%), as were the most common events (fatigue, rash, diarrhea, hypothyroidism, nausea, and pruritus). However, proportions experiencing 'significant' TRAEs varied between cohorts. Proportions discontinuing treatment were relatively consistent across cohorts. Propensity score matching and sensitivity analyses generally supported the unadjusted findings. CONCLUSIONS Real-world safety profiles of nivolumab 240 mg Q2W and 480 mg Q4W were similar, and both were comparable to the well-documented safety of weight-based dosing (3 mg/kg Q2W), further supporting their approval and use in the adjuvant setting for melanoma.
Collapse
Affiliation(s)
- Wolfram Samlowski
- Comprehensive Cancer Centers of Nevada and University of Nevada Las Vegas Kirkorian School of MedicineLas VegasNevadaUSA,University of Nevada School of MedicineRenoNevadaUSA
| | | | - Liwei Chen
- McKesson Life SciencesThe WoodlandsTexasUSA
| | | | | | | | | | | | - Jeffrey S. Weber
- Perlmutter Cancer CenterNYU School of MedicineNew YorkNew YorkUSA
| |
Collapse
|
31
|
Morimoto K, Yamada T, Morimoto Y, Ishikawa T, Asai J, Fujihara A, Arai A, Katoh N, Ukimura O, Hirano S, Itoh Y, Takayama K. A real-world study on the safety of the extended dosing schedule for nivolumab and pembrolizumab in patients with solid tumors. Int Immunopharmacol 2022; 108:108775. [DOI: 10.1016/j.intimp.2022.108775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/22/2022]
|
32
|
Jiang M, Hu Y, Lin G, Chen C. Dosing Regimens of Immune Checkpoint Inhibitors: Attempts at Lower Dose, Less Frequency, Shorter Course. Front Oncol 2022; 12:906251. [PMID: 35795044 PMCID: PMC9251517 DOI: 10.3389/fonc.2022.906251] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/24/2022] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a revolutionary breakthrough in the field of cancer by modulating patient's own immune system to exert anti-tumor effects. The clinical application of ICIs is still in its infancy, and their dosing regimens need to be continuously adjusted. Pharmacokinetic/pharmacodynamic studies showed a significant plateau in the exposure-response curve, with high receptor occupancy and plasma concentrations achieved at low dose levels. Coupled with concerns about drug toxicity and heavy economic costs, there has been an ongoing quest to reevaluate the current ICI dosing regimens while preserving maximum clinical efficacy. Many clinical data showed remarkable anticancer effects with ICIs at the doses far below the approved regimens, indicating the possibility of dose reduction. Our review attempts to summarize the clinical evidence for ICIs regimens with lower-dose, less-frequency, shorter-course, and provide clues for further ICIs regimen optimization.
Collapse
Affiliation(s)
| | | | | | - Chao Chen
- Department of Radiotherapy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
33
|
Teoh JY, Jeon S, Yim B, Yang HM, Hwang Y, Kim J, Lee SK, Park E, Kong TY, Kim SY, Park Y, Kim YG, Kim J, Yoo D. Tuning Surface Plasmon Resonance Responses through Size and Crosslinking Control of Multivalent Protein Binding-Capable Nanoscale Hydrogels. ACS Biomater Sci Eng 2022; 8:2878-2889. [PMID: 35658391 DOI: 10.1021/acsbiomaterials.2c00250] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Surface plasmon resonance (SPR) phenomena have been widely studied to detect biomolecules because of their high sensitivity and ability to determine biomolecular interactions with kinetic information. However, highly selective detection in specific concentration ranges relevant to target biomolecules is still a challenging task. Recently, we developed bioresponsive nanoscale hydrogels to selectively intensify SPR signals through multivalent protein binding (MPB) events with target biomolecules, including IL-2, where we were able to demonstrate exceptional selectivity for target biomolecules with minimal responses to nonspecific and monovalent binding events. In this work, we systematically explored the relationship between the physical properties of MPB-capable nanoscale hydrogels and their SPR response induced in the presence of the programmed cell death protein 1 antibody (PD-1Ab) as a model target biomolecule. First, we developed a synthetic protocol by controlling various reaction parameters to construct a library of nanoscale poly(N-isopropylacrylamide-co-acrylic acid) hydrogels (NHs) with different sizes (from 400 nm to 1 μm) and degrees of crosslinking (from 2 to 8%). Then, by incorporating MPB-capable PD-1 receptors onto the surface of NHs to form PD-1-responsive nanoscale hydrogels (PNHs), the hydrogel size and crosslinking dependency of their SPR responses were investigated. Our results reveal the appropriate hydrogel size regime and degree of crosslinking for effective PD-1Ab detection at specific concentrations range between a few nM and 1 μM. Overall, our study demonstrates that by tuning the physical properties of the nanoscale hydrogel matrix, the sensitivity and detection range of MPB-based SPR sensors can be modulated to potentially benefit clinical applications such as monitoring diverse therapeutic biomolecules.
Collapse
Affiliation(s)
- Jie Ying Teoh
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Suhwan Jeon
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Bora Yim
- R&D Center, Scholar Foxtrot Co. Ltd., Seoul 02841, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Hae Min Yang
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Yunseo Hwang
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Juhui Kim
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Su-Kyoung Lee
- R&D Center, Scholar Foxtrot Co. Ltd., Seoul 02841, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Eunyoung Park
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Tae Yeon Kong
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - So Youn Kim
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Young Gyu Kim
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea
| | - Jongseong Kim
- R&D Center, Scholar Foxtrot Co. Ltd., Seoul 02841, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Dongwon Yoo
- Department of Chemical and Biological Engineering, and Institute of Chemical Process, Seoul National University, Seoul 08826, Republic of Korea.,Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
| |
Collapse
|
34
|
Liu M, Que Y, Hong Y, Zhang L, Zhang X, Zhang Y. A Pan-Cancer Analysis of IRAK1 Expression and Their Association With Immunotherapy Response. Front Mol Biosci 2022; 9:904959. [PMID: 35669566 PMCID: PMC9163706 DOI: 10.3389/fmolb.2022.904959] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/29/2022] [Indexed: 11/14/2022] Open
Abstract
IRAK1 is an active kinase which plays a critical role in IL-1/TLR signaling pathway involved in inflammation and innate immune response. Recently, increasing evidence supports a potential role of IRAK1 in cancer progression. However, no immunological pan-cancer analysis of IRAK1 is available. We aimed to explore the prognostic value and the immunological functions of IRAK1. A series of datasets including The Cancer Genome Atlas, GEPIA2, cBioPortal, HPA, TIMER2.0 were performed to explore the oncogenic and immunological roles of IRAK1, including the relationship between IRAK1 and prognosis, genetic mutation, GO and KEGG enrichment pathway analysis, immune state of different tumors, The results showed that IRAK1 levels were upregulated in more than 20 types of cancers compared to the normal tissues. IRAK1 expression was associated with poorer prognosis in different cancer types. For the most frequent DNA alteration of IRAK1 is amplification. And the result of the enrichment analysis suggested that IRAK1 related to immune checkpoint pathway in cancer. IRAK1 inhibitor pacritinib inhibit proliferation and upregulate PD-L1 expression in different cancer cell lines. Moreover, the patients who receiving anti-PD-L1 therapy with low IRAK1 expression had a better prognosis, and the objective response rate to anti-PD-L1 therapy was higher in the low IRAK1 group than in the high IRAK1 group in IMvigor210 cohort. Our study reveals that IRAK1 can function as a prognostic marker in various malignant tumors. And pacritinib upregulated PD-L1 expression in several cancer cell lines, which indicating that IRAK1 can be used as a reliable marker to predict the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Mengmeng Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Melanoma and Sarcoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Que
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ye Hong
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lian Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Melanoma and Sarcoma, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Xing Zhang, ; Yizhuo Zhang,
| | - Yizhuo Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Xing Zhang, ; Yizhuo Zhang,
| |
Collapse
|
35
|
Hamuro L, Hu Z, Passarell J, Barcomb H, Zhang J, Goldstein S, Bello A, Roy A, Zhu L. Exposure-Response Analysis to Support Nivolumab Once Every 4 Weeks Dosing in Combination with Cabozantinib in Renal Cell Carcinoma. Clin Cancer Res 2022; 28:1603-1613. [PMID: 34980597 DOI: 10.1158/1078-0432.ccr-21-3149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/22/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE A benefit:risk assessment for a less-frequent nivolumab 480 mg Q4W + cabozantinib 40 mg QD dosing regimen was predicted using modeling and simulation of clinical trial data from nivolumab monotherapy studies and from the nivolumab 240 mg Q2W + cabozantinib 40mg QD dosing regimen, which demonstrated clinical benefit versus sunitinib in previously untreated advanced renal cell carcinoma (aRCC) in the phase III CheckMate 9ER trial (NCT03141177). EXPERIMENTAL DESIGN Multivariable Cox proportional-hazards analyses were conducted using nivolumab monotherapy data in previously treated aRCC and data from CheckMate 9ER to evaluate progression-free survival (PFS), overall survival (OS), and grade {greater than or equal to}2 immune-mediated adverse events (IMAEs). RESULTS Nivolumab 240 mg Q2W + cabozantinib versus nivolumab showed improvement in PFS (HR, 0.38; 95% CI, 0.31-0.47), OS (HR, 0.63 95% CI, 0.46-0.85), and increased risk of grade {greater than or equal to}2 IMAEs (HR, 2.19; 95% CI, 1.79-2.67). Nivolumab exposure was not a predictor of PFS/OS or grade {greater than or equal to}2 IMAEs. Lower nivolumab clearance, male sex, higher baseline bodyweight, and Karnofsky performance (100) were each associated with PFS/OS improvements. Region and IMDC poor score were negative OS predictors. Age, baseline albumin, and programmed death ligand-1 status were not significant PFS/OS predictors. Cabozantinib was a significant grade {greater than or equal to}2 IMAE predictor, driven by diarrhea and hepatic events. Model-predicted PFS/OS and grade {greater than or equal to}2 IMAE rates were similar (<2.5% difference) for nivolumab 240 mg Q2W + cabozantinib and 480 mg Q4W + cabozantinib. CONCLUSIONS Comparable benefit:risk was predicted for nivolumab 480 mg Q4W + cabozantinib and nivolumab 240 mg Q2W + cabozantinib.
Collapse
Affiliation(s)
- Lora Hamuro
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb (United States)
| | - Zheyi Hu
- Bristol-Myers Squibb (United States)
| | | | | | | | | | - Akintunde Bello
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb (United States)
| | - Amit Roy
- Clinical Pharmacology and Pharmacometrics, Research and Development, Bristol-Myers Squibb (United States)
| | - Li Zhu
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb (United States)
| |
Collapse
|
36
|
Hu J, Albadawi H, Zhang Z, Salomao MA, Gunduz S, Rehman S, D'Amone L, Mayer JL, Omenetto F, Oklu R. Silk Embolic Material for Catheter-Directed Endovascular Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106865. [PMID: 34695275 PMCID: PMC8758542 DOI: 10.1002/adma.202106865] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/13/2021] [Indexed: 05/06/2023]
Abstract
Embolization is a catheter-based minimally invasive procedure that deliberately occludes diseased blood vessels for treatment purposes. A novel silk-based embolic material (SEM) that is developed and optimized to provide tandem integration of both embolization and the delivery of therapeutics is reported. Natural silk is processed into fibroin proteins of varying lengths and is combined with charged nanoclay particles to allow visibility and injectability using clinical catheters as small as 600 μm in diameter at lengths >100 cm. SEMs loaded with fluorochrome labeled bovine albumin and Nivolumab, which is among the most used immunotherapy drugs worldwide, demonstrate a sustained release profile in vitro over 28 days. In a porcine renal survival model, SEMs with labeled albumin and Nivolumab successfully embolize porcine arteries without recanalization and lead to the delivery of both albumin and Nivolumab into the interstitial space of the renal cortex. Mechanistically, it is shown that tissue delivery is most optimal when the internal elastic membrane of the embolized artery is disrupted. SEM is a potential next-generation multifunctional embolic agent that can achieve embolization and deliver a wide range of therapeutics to treat vascular diseases including tumors.
Collapse
Affiliation(s)
- Jingjie Hu
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ, 85259, USA
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ, 85259, USA
| | - Zefu Zhang
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ, 85259, USA
| | - Marcela A Salomao
- Division of Anatomic Pathology and Laboratory Medicine, Department of Pathology, Mayo Clinic, 5777 East Mayo Blvd., Phoenix, AZ, 85054, USA
| | - Seyda Gunduz
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ, 85259, USA
| | - Suliman Rehman
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ, 85259, USA
| | - Luciana D'Amone
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
| | - Joseph L Mayer
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ, 85259, USA
| | - Fiorenzo Omenetto
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA, 02155, USA
- Laboratory for Living Devices, Tufts University, Medford, MA, 02155, USA
- Department of Electrical and Computer Engineering, Tufts University, Medford, MA, 02155, USA
- Department of Physics, Tufts University, Medford, MA, 02155, USA
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, 13400 East Shea Blvd., Scottsdale, AZ, 85259, USA
| |
Collapse
|
37
|
Safety and Efficacy of Extended Interval Dosing for Immune Checkpoint Inhibitors in Non–Small Cell Lung Cancer During the COVID-19 Pandemic. Clin Lung Cancer 2021; 23:143-150. [PMID: 35034861 PMCID: PMC8704727 DOI: 10.1016/j.cllc.2021.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 11/21/2022]
Abstract
Introduction Extended interval (EI) dosing for immune checkpoint inhibitor (ICI) mono- or consolidation therapy initiated due to the COVID-19 pandemic led to a significant reduction in ICI-related site visits for patients with stage III and IV non–small cell lung cancer. Here we report the safety and efficacy compared to standard dose (SD) schedules. Method In this retrospective analysis, patients who received ICI mono- or consolidation therapy, or adjuvant ICI therapy were assessed. Safety and efficacy of EI dosing with data of SD schedules were compared. Results One hundred seventeen patients received EI dosing for ICI and 88 patients SD. Patient characteristics were comparable. We observed 237 adverse events in the EI dosing cohort versus 118 in the SD group (P= .02). Overall, there was no difference in the occurrence of grade ≥3 adverse events (EI dosing: 21/237 [8.9%]; SD group: 20/118 [17.0%], P = .42), except for the pembrolizumab EI dosing cohort. Of all patients who received an EI dosing schedule, however, only 8 (6.8%) were reduced to SD because of toxicity. In 5 (4.3%) patients ICI was permanently stopped because of severe toxicity compared to 11 (12.5%) discontinuations in the SD group. Short-term treatment interruption occurred with similar frequencies in both groups. Progression-free survival and overall survival were comparable in patients receiving pembrolizumab and in those receiving adjuvant durvalumab. Progression-free survival and OS were better in the EI dosing cohort of nivolumab. Conclusion EI dosing for ICI did not lead to an increase of clinically relevant toxicities resulting in dose reduction and/or treatment discontinuation. Efficacy of EI dosing of pembrolizumab and durvalumab were comparable to SD. Based on our safety and efficacy data EI dosing for ICI seems a safe and effective strategy.
Collapse
|
38
|
Gulikers JL, Croes S, Schreurs MJW, Litjens EJR, Aarts MJB, van Geel RMJM. Nivolumab exposure in a hemodialysis patient with metastatic melanoma. Melanoma Res 2021; 31:579-581. [PMID: 34433199 DOI: 10.1097/cmr.0000000000000775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The effect of intermittent hemodialysis (IHD) on nivolumab serum concentrations in patients with severe renal impairment is largely unknown. Here, we present a 79-year-old patient with metastatic melanoma and end-stage renal disease on IHD three times a week, treated with 480 mg nivolumab every 4 weeks. A serum trough concentration of nivolumab was determined before the start of the third cycle, and two samples were taken immediately before and after a hemodialysis session during this cycle. All nivolumab serum concentrations were within a similar range as those previously measured among patients without renal insufficiency, after a comparable duration of nivolumab treatment. Therefore, we conclude that IHD does not influence nivolumab exposure. Furthermore, nivolumab treatment was continued without complications and appears to be well tolerated for patients on IHD.
Collapse
Affiliation(s)
- Judith L Gulikers
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht
| | - Sander Croes
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht
| | - Marco J W Schreurs
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam
| | - Elisabeth J R Litjens
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center
| | - Maureen J B Aarts
- Department of Medical Oncology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Robin M J M van Geel
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht
| |
Collapse
|
39
|
Cao Y, Chen C, Tao Y, Lin W, Wang P. Immunotherapy for Triple-Negative Breast Cancer. Pharmaceutics 2021; 13:2003. [PMID: 34959285 PMCID: PMC8705248 DOI: 10.3390/pharmaceutics13122003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/03/2021] [Accepted: 11/23/2021] [Indexed: 01/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by extensive tumor heterogeneity at both the pathologic and molecular levels, particularly accelerated aggressiveness, and terrible metastasis. It is responsible for the increased mortality of breast cancer patients. Due to the negative expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2, the progress of targeted therapy has been hindered. Higher immune response in TNBCs than for other breast cancer types makes immunotherapy suitable for TNBC therapy. At present, promising treatments in immunotherapy of TNBC include immune checkpoints (ICs) blockade therapy, adoptive T-cell immunotherapy, and tumor vaccine immunotherapy. In addition, nanomedicines exhibit great potential in cancer therapy through the enhanced permeability and retention (EPR) effect. Immunotherapy-involved combination therapy may exert synergistic effects by combining with other treatments, such as traditional chemotherapy and new treatments, including photodynamic therapy (PTT), photodynamic therapy (PDT), and sonodynamic therapy (SDT). This review focuses on introducing the principles and latest development as well as progress in using nanocarriers as drug-delivery systems for the immunotherapy of TNBC.
Collapse
Affiliation(s)
- Yifeng Cao
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (C.C.); (Y.T.)
| | - Chuyang Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (C.C.); (Y.T.)
| | - Yi Tao
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (C.C.); (Y.T.)
| | - Weifeng Lin
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Ping Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China; (C.C.); (Y.T.)
| |
Collapse
|
40
|
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
|
41
|
Travert C, Cannone P, Greillier L, Tomasini P. [Immunotherapy adaptation in lung cancer during the COVID-19 pandemic]. Rev Mal Respir 2021; 38:865-872. [PMID: 34246519 PMCID: PMC8241593 DOI: 10.1016/j.rmr.2021.06.005] [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: 03/25/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION COVID-19 pandemics required changes in medical practices. In thoracic oncology, pembrolizumab was doubled to 400mg every 6weeks, nivolumab to 480mg every 4weeks. The objective of our study was to assess the impact on quality of life, and on psychological state, as well as the tolerance, of this new schedule. METHODS Thoracic oncologic patients who underwent these therapeutic changes in our center during the first COVID-19 epidemic wave were included. Their quality of life was assessed using the Quality of Life Questionnaire-30, their psychological state by the Hospital Anxiety Depression (HAD) scale. We also reported the preferred administration schedule, as well as adverse events. RESULTS Thirty patients were included. The overall quality of life was preserved. Rates on HAD scale were low. Tolerance was acceptable. In majority, patients preferred the new procedure. They had a significantly better quality of life compared to those who preferred the old one. CONCLUSIONS This new immunotherapy schedule in thoracic oncology is well tolerated and allows a preservation of quality of life. This therapeutic option may be favored in the context of COVID-19 pandemics.
Collapse
Affiliation(s)
- C Travert
- Aix-Marseille université, AP-HM, INSERM, CNRS, CRCM, hôpital Nord, service d'oncologie multidisciplinaire et innovations thérapeutiques, Marseille, France; Institut du cancer de Montpellier, Montpellier, France
| | - P Cannone
- Aix-Marseille université, AP-HM, INSERM, CNRS, CRCM, hôpital Nord, service d'oncologie multidisciplinaire et innovations thérapeutiques, Marseille, France
| | - L Greillier
- Aix-Marseille université, AP-HM, INSERM, CNRS, CRCM, hôpital Nord, service d'oncologie multidisciplinaire et innovations thérapeutiques, Marseille, France
| | - P Tomasini
- Aix-Marseille université, AP-HM, INSERM, CNRS, CRCM, hôpital Nord, service d'oncologie multidisciplinaire et innovations thérapeutiques, Marseille, France.
| |
Collapse
|
42
|
Zhou S, Sun Y, Chen T, Wang J, He J, Lyu J, Shen Y, Chen X, Yang R. The Landscape of the Tumor Microenvironment in Skin Cutaneous Melanoma Reveals a Prognostic and Immunotherapeutically Relevant Gene Signature. Front Cell Dev Biol 2021; 9:739594. [PMID: 34660598 PMCID: PMC8517264 DOI: 10.3389/fcell.2021.739594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/23/2021] [Indexed: 01/15/2023] Open
Abstract
The tumorigenesis of skin cutaneous melanoma (SKCM) remains unclear. The tumor microenvironment (TME) is well known to play a vital role in the onset and progression of SKCM. However, the dynamic mechanisms of immune regulation are insufficient. We conducted a comprehensive analysis of immune cell infiltration in the TME. Based on the differentially expressed genes (DEGs) in clusters grouped by immune infiltration status, a set of hub genes related to the clinical prognosis of SKCM and tumor immune infiltration was explored. Methods: We analyzed immune cell infiltration in two independent cohorts and assessed the relationship between the internal pattern of immune cell infiltration and SKCM characteristics, including clinicopathological features, potential biological pathways, and gene mutations. Genes related to the infiltration pattern of TME immune cells were determined. Furthermore, the unsupervised clustering method (k-means) was used to divide samples into three different categories according to TME, which were defined as TME cluster-A, -B, and -C. DEGs among three groups of samples were analyzed as signature genes. We further distinguished common DEGs between three groups of samples according to whether differences were significant and divided DEGs into the Signature gene-A group with significant differences and the Signature gene-B group with insignificant differences. The Signature gene-A gene set mainly had exon skipping in SKCM, while the Signature gene-B gene set had no obvious alternative splicing form. Subsequently, we analyzed genetic variations of the two signatures and constructed a competing endogenous RNA (ceRNA) regulatory network. LASSO Cox regression was used to determine the immune infiltration signature and risk score of SKCM. Finally, we obtained 13 hub genes and calculated the risk score based on the coefficient of each gene to explore the impact of the high- and low-risk scores on biologically related functions and prognosis of SKCM patients further. The correlation between the risk score and clinicopathological characteristics of SKCM patients indicated that a low-risk score was associated with TME cluster-A classification (p < 0.001) and metastatic SKCM (p < 0.001). Thirteen hub genes also showed different prognostic effects in pan-cancer. The results of univariate and multivariate Cox analyses revealed that risk score could be used as an independent risk factor for predicting the prognosis of SKCM patients. The nomogram that integrated clinicopathological characteristics and immune characteristics to predict survival probability was based on multivariate Cox regression. Finally, 13 hub genes that showed different prognostic effects in pan-cancers were obtained. According to immunohistochemistry staining results, Ube2L6, SRPX2, and IFIT2 were expressed at higher levels, while CLEC4E, END3, and KIR2DL4 were expressed at lower levels in 25 melanoma specimens. Conclusion: We performed a comprehensive assessment of the immune-associated TME. To elucidate the potential development of immune-genomic features in SKCM, we constructed an unprecedented set of immune characteristic genes (EDN3, CLEC4E, SRPX2, KIR2DL4, UBE2L6, and IFIT2) related to the immune landscape of TME. These genes are related to different prognoses and drug responses of SKCM. The immune gene signature constructed can be used as a robust prognostic biomarker of SKCM and a predictor of an immunotherapy effect.
Collapse
Affiliation(s)
- Sitong Zhou
- Department of Dermatology, The First People's Hospital of Foshan, Foshan, China
| | - Yidan Sun
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tianqi Chen
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingru Wang
- Department of Burn Surgery and Skin Regeneration, The First People's Hospital of Foshan, Foshan, China
| | - Jia He
- Department of Burn Surgery and Skin Regeneration, The First People's Hospital of Foshan, Foshan, China
| | - Jin Lyu
- Department of Pathology, The First People's Hospital of Foshan, Foshan, China
| | - Yanna Shen
- School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xiaodong Chen
- Department of Burn Surgery and Skin Regeneration, The First People's Hospital of Foshan, Foshan, China
| | - Ronghua Yang
- Department of Burn Surgery and Skin Regeneration, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
43
|
Bobircă A, Bobircă F, Ancuta I, Florescu A, Pădureanu V, Florescu DN, Pădureanu R, Florescu A, Mușetescu AE. Rheumatic Immune-Related Adverse Events-A Consequence of Immune Checkpoint Inhibitor Therapy. BIOLOGY 2021; 10:561. [PMID: 34203101 PMCID: PMC8235766 DOI: 10.3390/biology10060561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/04/2022]
Abstract
The advent of immunotherapy has changed the management and therapeutic methods for a variety of malignant tumors in the last decade. Unlike traditional cytotoxic chemotherapy, which works by interfering with cancer cell growth via various pathways and stages of the cell cycle, cancer immunotherapy uses the immune system to reduce malignant cells' ability to escape the immune system and combat cell proliferation. The widespread use of immune checkpoint inhibitors (ICIs) over the past 10 years has presented valuable information on the profiles of toxic adverse effects. The attenuation of T-lymphocyte inhibitory mechanisms by ICIs results in immune system hyperactivation, which, as expected, is associated with various adverse events defined by inflammation. These adverse events, known as immune-related adverse events (ir-AEs), may affect any type of tissue throughout the human body, which includes the digestive tract, endocrine glands, liver and skin, with reports of cardiovascular, pulmonary and rheumatic ir-AEs as well. The adverse events that arise from ICI therapy are both novel and unique compared to those of the conventional treatment options. Thus, they require a multidisciplinary approach and continuous updates on the diagnostic approach and management.
Collapse
Affiliation(s)
- Anca Bobircă
- Department of Internal Medicine and Rheumatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.B.); (I.A.)
| | - Florin Bobircă
- Department of General Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Ioan Ancuta
- Department of Internal Medicine and Rheumatology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.B.); (I.A.)
| | - Alesandra Florescu
- Department of Rheumatology, Emergency Clinical County Hospital of Craiova, 200642 Craiova, Romania;
| | - Vlad Pădureanu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Dan Nicolae Florescu
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Rodica Pădureanu
- Department of Internal Medicine, Emergency Clinical County Hospital of Craiova, 200642 Craiova, Romania
| | - Anca Florescu
- Department of Internal Medicine and Rheumatology, Dr. I Cantacuzino Hospital, 030167 Bucharest, Romania;
| | - Anca Emanuela Mușetescu
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
44
|
Hyytiäinen A, Wahbi W, Väyrynen O, Saarilahti K, Karihtala P, Salo T, Al-Samadi A. Angiogenesis Inhibitors for Head and Neck Squamous Cell Carcinoma Treatment: Is There Still Hope? Front Oncol 2021; 11:683570. [PMID: 34195084 PMCID: PMC8236814 DOI: 10.3389/fonc.2021.683570] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/17/2021] [Indexed: 01/27/2023] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) carries poor survival outcomes despite recent progress in cancer treatment in general. Angiogenesis is crucial for tumour survival and progression. Therefore, several agents targeting the pathways that mediate angiogenesis have been developed. We conducted a systematic review to summarise the current clinical trial data examining angiogenesis inhibitors in HNSCC. Methods We carried out a literature search on three angiogenesis inhibitor categories—bevacizumab, tyrosine kinase inhibitors and endostatin—from Ovid MEDLINE, Cochrane Library, Scopus and ClinicalTrials.gov database. Results Here, we analysed 38 clinical trials, total of 1670 patients, investigating 12 angiogenesis inhibitors. All trials were in phase I or II, except one study in phase III on bevacizumab. Angiogenesis inhibitors were used as mono- and combination therapies together with radio-, chemo-, targeted- or immunotherapy. Among 12 angiogenesis inhibitors, bevacizumab was the most studied drug, included in 13 trials. Although bevacizumab appeared effective in various combinations, it associated with high toxicity levels. Endostatin and lenvatinib were well-tolerated and their anticancer effects appeared promising. Conclusions Most studies did not show benefit of angiogenesis inhibitors in HNSCC treatment. Additionally, angiogenesis inhibitors were associated with considerable toxicity. However, some results appear encouraging, suggesting that further investigations of angiogenesis inhibitors, particularly in combination therapies, for HNSCC patients are warranted. Systematic Review Registration PROSPERO (https://www.crd.york.ac.uk/prospero/), identifier CRD42020157144.
Collapse
Affiliation(s)
- Aini Hyytiäinen
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Otto Väyrynen
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Kauko Saarilahti
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Centre and University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Pathology, University of Helsinki, Helsinki, Finland.,Cancer Research and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Oulu Medical Research Centre, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland.,Translational Immunology Programme, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
45
|
Schlicke P, Kuttler C, Schumann C. How mathematical modeling could contribute to the quantification of metastatic tumor burden under therapy: insights in immunotherapeutic treatment of non-small cell lung cancer. Theor Biol Med Model 2021; 18:11. [PMID: 34078405 PMCID: PMC8170801 DOI: 10.1186/s12976-021-00142-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cancer is one of the leading death causes globally with about 8.2 million deaths per year and an increase in numbers in recent years. About 90% of cancer deaths do not occur due to primary tumors but due to metastases, of which most are not clinically identifiable because of their relatively small size at primary diagnosis and limited technical possibilities. However, therapeutic decisions are formed depending on the existence of metastases and their properties. Therefore non-identified metastases might have huge influence in the treatment outcome. The quantification of clinically visible and invisible metastases is important for the choice of an optimal treatment of the individual patient as it could clarify the burden of non-identifiable tumors as well as the future behavior of the cancerous disease. RESULTS The mathematical model presented in this study gives insights in how this could be achieved, taking into account different treatment possibilities and therefore being able to compare therapy schedules for individual patients with different clinical parameters. The framework was tested on three patients with non-small cell lung cancer, one of the deadliest types of cancer worldwide, and clinical history including platinum-based chemotherapy and PD-L1-targeted immunotherapy. Results yield promising insights into the framework to establish methods to quantify effects of different therapy methods and prognostic features for individual patients already at stage of primary diagnosis.
Collapse
Affiliation(s)
- Pirmin Schlicke
- Center of Mathematics, Technical University of Munich, Boltzmannstraße, Garching, Germany.
| | - Christina Kuttler
- Center of Mathematics, Technical University of Munich, Boltzmannstraße, Garching, Germany
| | - Christian Schumann
- Clinic of Pneumology, Thoracic Oncology, Sleep and Respiratory Critical Care, Klinikverbund Allgäu, Robert-Weichsler-Straße, Kempten, Germany
| |
Collapse
|
46
|
Silvestris N, Argentiero A, Natalicchio A, D'Oronzo S, Beretta GD, Acquati S, Adinolfi V, Di Bartolo P, Danesi R, Faggiano A, Ferrari P, Gallo M, Gori S, Morviducci L, Russo A, Tuveri E, Zatelli MC, Montagnani M, Giorgino F. Antineoplastic dosing in overweight and obese cancer patients: an Associazione Italiana Oncologia Medica (AIOM)/Associazione Medici Diabetologi (AMD)/Società Italiana Endocrinologia (SIE)/Società Italiana Farmacologia (SIF) multidisciplinary consensus position paper. ESMO Open 2021; 6:100153. [PMID: 33984679 PMCID: PMC8134762 DOI: 10.1016/j.esmoop.2021.100153] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/23/2021] [Accepted: 04/14/2021] [Indexed: 01/22/2023] Open
Abstract
Most anticancer molecules are administered in body-size-based dosing schedules, bringing up unsolved issues regarding pharmacokinetic data in heavy patients. The worldwide spread of obesity has not been matched by improved methods and strategies for tailored drug dosage in this population. The weight or body surface area (BSA)-based approaches may fail to fully reflect the complexity of the anthropometric features besides obesity in cancer patients suffering from sarcopenia. Likewise, there is a lack of pharmacokinetic data on obese patients for the majority of chemotherapeutic agents as well as for new target drugs and immunotherapy. Therefore, although the available findings point to the role of dose intensity in cancer treatment, and support full weight-based dosing, empirical dose capping often occurs in clinical practice in order to avoid toxicity. Thus a panel of experts of the Associazione Italiana Oncologia Medica (AIOM), Associazione Medici Diabetologi (AMD), Società Italiana Endocrinologia (SIE), and Società Italiana Farmacologia (SIF), provides here a consensus statement for appropriate cytotoxic chemotherapy and new biological cancer drug dosing in obese patients. The worldwide spread of obesity is an emerging challenge also in cancer patients Weight or BSA-based approaches do not adequately address the critical issue of optimal dosing for cancer drugs under obesity Empirical dose capping is often employed in clinical practice to avoid toxicities among overweight and obese patients There is a lack of clinical and pharmacokinetic studies in this population Clinical practice recommendations should guide suitable dosing of cytotoxic and biological cancer drugs in obese patients
Collapse
Affiliation(s)
- N Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori 'Giovanni Paolo II', Bari, Italy; Department of Biomedical Sciences and Human Oncology, Division of Medical Oncology, University of Bari Aldo Moro, Bari, Italy.
| | - A Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - A Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - S D'Oronzo
- Department of Biomedical Sciences and Human Oncology, Division of Medical Oncology, University of Bari Aldo Moro, Bari, Italy; Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
| | - G D Beretta
- Medical Oncology Department, Humanitas Gavazzeni, Bergamo, Italy
| | - S Acquati
- Endocrinology Unit, Ospedale Pierantoni-Morgagni, Forlì, Italy
| | - V Adinolfi
- Endocrinology and Diabetology Unit, ASL Verbano Cusio Ossola, Domodossola, Italy
| | - P Di Bartolo
- Diabetology Clinic, Rete Clinica di Diabetologia Aziendale - Dipartimento, Internistico di Ravenna - AUSL Romagna, Ravenna, Italy
| | - R Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A Faggiano
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - P Ferrari
- Palliative Care Unit, Istituti Clinici Scientifici Maugeri SPA SB, IRCCS (PV), Pavia PV, Italy
| | - M Gallo
- Endocrinology and Metabolic Diseases Unit of AO SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - S Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona, Italy
| | - L Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialities, ASL Roma 1 - S. Spirito Hospital, Rome, Italy
| | - A Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - E Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ATS Sardegna - ASSL Carbonia-Iglesias, Italy
| | - M C Zatelli
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - M Montagnani
- Department of Biomedical Sciences and Human Oncology, Division of Medical Oncology, University of Bari Aldo Moro, Bari, Italy
| | - F Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
47
|
Sanghavi K, Vuppala P, Ivaturi V, Hamuro L, Roy A, Suryawanshi S. Nivolumab exposure-response analysis for adjuvant treatment of melanoma supporting a change in posology. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:748-759. [PMID: 33955714 PMCID: PMC8302247 DOI: 10.1002/psp4.12642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/11/2021] [Accepted: 04/01/2021] [Indexed: 01/05/2023]
Abstract
Nivolumab monotherapy is approved as adjuvant treatment for melanoma based on results from the pivotal CheckMate 238 trial. We present a model‐based, benefit–risk assessment of nivolumab in adjuvant melanoma supporting a posology change from a weight‐based to a less frequent, flat‐dosing regimen. The exposure–response (E–R) relationship for efficacy was evaluated using recurrence‐free survival (RFS) and distant metastasis‐free survival (DMFS) end points from the CheckMate 238 trial. The E–R for safety was evaluated using data from 14 studies across a broad range of doses in several tumor types using grade 3+ adverse event (AE) and grade 2+ immune‐mediated AE (IMAE) end points. Nivolumab trough exposures were not significant predictors of RFS or DMFS. Covariates significantly associated with increased risk of disease recurrence or death were programmed death ligand 1 (PD‐L1; less than 5% cutoff), lower baseline lactate dehydrogenase, and higher age. Covariates associated with increased risk of distant metastasis or death were PD‐L1 (less than 5% cutoff) and higher age. Higher nivolumab maximum concentration after first dose (Cmax1) was significantly associated with grade 2+ IMAEs, but not grade 3+ AEs. The risk of grade 3+ AEs was significantly lower in adjuvant versus advanced melanoma. Eastern Cooperative Oncology Group Performance Status higher than zero was associated with higher incidences of grade 2+ IMAEs and grade 3+ AEs. Female patients had significantly higher incidences of grade 2+ IMAEs than male patients. Nivolumab monotherapy in adjuvant melanoma demonstrated a relatively flat E–R relationship over the range of exposures produced by 3 mg/kg every 2 weeks and predicted a comparable benefit–risk profile to flat‐dosing regimens.
Collapse
Affiliation(s)
| | | | | | - Lora Hamuro
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Amit Roy
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | |
Collapse
|
48
|
Evaluating the efficacy and safety of immune checkpoint inhibitors by detecting the exposure-response: An inductive review. Int Immunopharmacol 2021; 97:107703. [PMID: 33933843 DOI: 10.1016/j.intimp.2021.107703] [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: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 11/23/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have been demonstrated an effective treatment in multiple tumor type, which restore the immune response to against cancer cell. Currently, approved ICIs include anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4); anti-programmed cell death 1 (PD-1) and anti-programmed cell death ligand 1 (PD-L1) monoclonal antibodies (mAbs). In most these drugs, unique pharmacokinetic (PK) and pharmacodynamics (PD) have shown significant influence on clinical outcomes, which occurred by target-mediated drug concentration and time-varying drug clearance. An exposure-response (E-R) relationship has been used to describe the safety and efficacy of ICIs, and shown a plateaued E-R and time dependent changes in exposure. Using an enzyme linked immunosorbent assay (ELISA) or LC-MS/MS method to measure the peak concentration, trough concentration or area under the curve (AUC) of ICIs to assess the drug exposure. There are lots of covariates that have an influence on exposure, such as sex, clearance, body weight and tumor burden. In this review, we pooled data from studies of concentration or other pharmacokinetics parameter of mAbs to assess E-R in efficacy and safety.
Collapse
|
49
|
Tang Y, Li X, Cao Y. Which factors matter the most? Revisiting and dissecting antibody therapeutic doses. Drug Discov Today 2021; 26:1980-1990. [PMID: 33895315 DOI: 10.1016/j.drudis.2021.04.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/28/2021] [Accepted: 04/16/2021] [Indexed: 01/22/2023]
Abstract
Factors such as antibody clearance and target affinity can influence antibodies' effective doses for specific indications. However, these factors vary considerably across antibody classes, precluding direct and quantitative comparisons. Here, we apply a dimensionless metric, the therapeutic exposure affinity ratio (TEAR), which normalizes the therapeutic doses by antibody bioavailability, systemic clearance and target-binding property to enable direct and quantitative comparisons of therapeutic doses. Using TEAR, we revisited and dissected the doses of up to 60 approved antibodies. We failed to detect a significant influence of target baselines, turnovers or anatomical locations on antibody therapeutic doses, challenging the traditional perceptions. We highlight the importance of antibodies' modes of action for therapeutic doses and dose selections; antibodies that work through neutralizing soluble targets show higher TEARs than those working through other mechanisms. Overall, our analysis provides insights into the factors that influence antibody doses, and the factors that are crucial for antibodies' pharmacological effects.
Collapse
Affiliation(s)
- Yu Tang
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaobing Li
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
50
|
Pous A, Izquierdo C, Cucurull M, Sánchez S, Lezcano C, Domenech M, Llobera L, Plaja A, Moran T. Immune-checkpoint inhibitors for lung cancer patients amid the COVID-19 pandemic: a case report of severe meningoencephalitis after switching to an extended-interval higher flat-dose nivolumab regimen. Transl Lung Cancer Res 2021; 10:1917-1923. [PMID: 34012801 PMCID: PMC8107731 DOI: 10.21037/tlcr-20-1315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alternative dosage regimens for some anticancer therapies have been proposed in the midst of the SARS-COV-2 pandemic in order to protect the patients from attending to health care facilities. Flat-dosing of several immune-checkpoint inhibitors (ICIs), including nivolumab, have been established. Although generally well tolerated with no new safety signals, new dosages can associate novel individual toxicities. As the use of ICIs is increasing in cancer patients, the present case report is a reminder for clinicians of potential novel toxicities, as well as the need for an interdisciplinary approach for their recognition and treatment. We report the occurrence of a severe neurologic toxicity in a patient with non-small cell lung cancer (NSCLC) who developed should be changed to which occurred after two doses of extended higher interval flat-dose nivolumab despite two years of clinical stability on prior nivolumab regimen. Patient developed fever, language impairment and altered mental status. The work-up tests excluded other potential causes and the most likely diagnosis was meningoencephalitis. Fortunately, with medical treatment, which consisted of high dose steroids, the patient recovered to his baseline situation and symptoms did not recurred, even though nivolumab was resumed. Alternate ICI regimens may have unique immune-related adverse event profiles.
Collapse
Affiliation(s)
- Anna Pous
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Cristina Izquierdo
- Neurology Department. Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Marc Cucurull
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,Institut Germans Trias i Pujol, Barcelona, Spain.,Badalona Applied Research Group in Oncology, Barcelona, Spain
| | - Silvia Sánchez
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Clara Lezcano
- Pharmacy Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Marta Domenech
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,Institut Germans Trias i Pujol, Barcelona, Spain.,Badalona Applied Research Group in Oncology, Barcelona, Spain
| | - Laia Llobera
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Andrea Plaja
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Teresa Moran
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,Institut Germans Trias i Pujol, Barcelona, Spain.,Badalona Applied Research Group in Oncology, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona (UAB), Campus Can Ruti, Barcelona, Spain
| |
Collapse
|