1
|
Zuo P, Bonate P, Garg A, Matsangou M, Tang M. Population Pharmacokinetic Modeling and Exposure-Response Analysis for the Antibody-Drug Conjugate Enfortumab Vedotin in Locally Advanced or Metastatic Urothelial Carcinoma. Clin Pharmacol Ther 2024; 116:1278-1288. [PMID: 39039635 DOI: 10.1002/cpt.3383] [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: 12/20/2023] [Accepted: 06/24/2024] [Indexed: 07/24/2024]
Abstract
Enfortumab vedotin is a fully human monoclonal antibody directed to Nectin-4 and conjugated to monomethyl auristatin E (MMAE), approved for treatment of previously treated locally advanced or metastatic urothelial carcinoma (mUC). This population analysis characterized pharmacokinetics of enfortumab vedotin and free (unconjugated) MMAE, identified covariates affecting pharmacokinetics, and evaluated weight-based dosing for enfortumab vedotin. Exposure-response analyses characterized relationships between enfortumab vedotin and free MMAE exposures and efficacy/safety endpoints. Data from 748 patients with locally advanced or mUC in 5 clinical studies were analyzed using nonlinear mixed-effects modeling. Patients received enfortumab vedotin 0.50-1.25 mg/kg every 3 weeks or on days 1, 8, and 15 of a 28-day cycle. Relevant covariates retained in final models were evaluated for clinical relevance to enfortumab vedotin and free MMAE exposures. Although some covariates produced differences in exposure, the magnitude of changes was not clinically meaningful. Simulations indicated weight-based dosing yielded more consistent exposures across body weight groups vs. a hypothetical fixed-dose regimen of enfortumab vedotin 95 mg (calculated for median body weight, 75 kg). Exposure-response analysis showed average enfortumab vedotin concentrations were not a statistically significant predictor of overall survival (hazard ratio 0.91, 95% confidence interval: 0.72-1.14; P = 0.41); all exposure quartiles had a greater median overall survival than chemotherapy (11.0-12.6 vs. 9.0 months). Enfortumab vedotin and free MMAE exposures were statistically significant predictors of grade ≥ 3 treatment-related adverse events (both P < 0.0001). This analysis supports enfortumab vedotin 1.25 mg/kg on days 1, 8, and 15 of a 28-day cycle.
Collapse
Affiliation(s)
- Peiying Zuo
- Astellas Pharma, Inc., Northbrook, Illinois, USA
- Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA
| | - Peter Bonate
- Astellas Pharma, Inc., Northbrook, Illinois, USA
| | - Amit Garg
- Pfizer Inc., South San Francisco, California, USA
| | | | - Mei Tang
- Astellas Pharma, Inc., Northbrook, Illinois, USA
| |
Collapse
|
2
|
Xu C, Xin K, Kosloski MP, Butler A, Goulaouic H, Nivens MC, Kanamaluru V. Pharmacokinetics of Subcutaneous Itepekimab Injection With an Autoinjector Device and Prefilled Syringe in Healthy Participants. Clin Pharmacol Drug Dev 2024; 13:1181-1188. [PMID: 39308293 DOI: 10.1002/cpdd.1466] [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: 03/02/2024] [Accepted: 08/13/2024] [Indexed: 11/06/2024]
Abstract
Itepekimab, a monoclonal antibody against interleukin-33, has demonstrated clinical utility in previous studies in patients with asthma and chronic obstructive pulmonary disease. An autoinjector (AI) has been developed for administering itepekimab to facilitate further development. This study compared pharmacokinetics of single 300-mg itepekimab subcutaneous administration via an AI versus a prefilled syringe (PFS). Of 90 healthy volunteers enrolled in this Phase 1, parallel-design, randomized study and stratified by body weight (50 to <70 kg, ≥70 to <80 kg, ≥80 to 100 kg) and injection site (abdomen, thigh, or arm), 84 completed the study. Systemic exposure of itepekimab was similar for both groups. Point estimates for geometric mean ratios of pharmacokinetic parameters for AI versus PFS groups were 1.01 for maximum serum concentration, 1.06 for area under the serum concentration-time curve to the last quantifiable concentration, and 1.04 for area under the serum concentration-time curve extrapolated to infinity. The exposure was similar for both devices in each body weight and injection site subgroup. Overall, systemic exposure of 300-mg single-dose itepekimab in healthy participants was comparable when administered subcutaneously via an AI device and PFS, with an acceptable safety profile in both device groups.
Collapse
|
3
|
Rousseau A, Géraud A, Geiss R, Farcet A, Spano JP, Hamy AS, Gougis P. Safety of solid oncology drugs in older patients: a narrative review. ESMO Open 2024; 9:103965. [PMID: 39481329 DOI: 10.1016/j.esmoop.2024.103965] [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: 07/09/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
The older population represents ∼50%-60% of the population of newly diagnosed patients with cancer. Due to physiological and pathological aging and the increased presence of comorbidities and frailty factors, this population is at higher risk of serious toxicity from anticancer drugs and, consequently, often under-treated. Despite the complexity of these treatments, a good knowledge of the pharmacology of anticancer drugs and potentially risky situations can limit the emergence of potentially lethal toxicities in this population. This review focuses on optimizing systemic oncology treatments for older patients, emphasizing the unique characteristics of each therapeutic class and the necessity for a precautionary approach for this vulnerable population.
Collapse
Affiliation(s)
- A Rousseau
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - A Géraud
- Department of Medical Oncology, Institut Paoli-Calmette, Marseille, France
| | - R Geiss
- Department of Medical Oncology, Institut Curie, Université Paris Cité, Paris, France
| | - A Farcet
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - J-P Spano
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | - A-S Hamy
- Department of Medical Oncology, Institut Curie, Université Paris Cité, Paris, France; Residual Tumor and Response to Treatment, RT2Lab, INSERM, U932 Cancer & Immunity, Institut Curie, Université Paris Sciences Lettres, Paris, France
| | - P Gougis
- Department of Medical Oncology, Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France; Residual Tumor and Response to Treatment, RT2Lab, INSERM, U932 Cancer & Immunity, Institut Curie, Université Paris Sciences Lettres, Paris, France; Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance Publique - Hôpitaux de Paris (AP-HP), Centre d'Investigation Clinique (CIC-1901), Pharmacology Department, Pitié-Salpêtrière Hospital, Paris, France.
| |
Collapse
|
4
|
Na JY, Jeon J, Huh KY, Yu KS, Lee S, Eom J, Ahn J, You WK, Oh J. Population pharmacokinetic model of ABL001/CTX-009 (anti-VEGF/DLL4) in adult cancer patients with solid tumor. Cancer Sci 2024. [PMID: 39375952 DOI: 10.1111/cas.16363] [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: 01/08/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
ABL001/CTX-009 is a bispecific antibody targeting delta-like ligand-4 and vascular endothelial growth factor A. In this study, we developed a population pharmacokinetic (PK) model of ABL001/CTX-009 in patients with solid tumors. A total of 712 plasma concentrations from 30 patients with relapsed or refractory solid tumors were collected from a phase 1 study (NCT03292783). A population PK model was developed using a nonlinear mixed-effect method and was evaluated by graphical and numerical methods. Using the model, the steady-state concentrations were simulated to compare weight-based and fixed-dose regimens and to find optimal dosing intervals. The PK of ABL001/CTX-009 was well described by a two-compartment model with a parallel first-order and Michaelis-Menten elimination kinetics. Body weight was selected as a significant covariate on V1. Model evaluation results suggested that the model was adequate and robust with good precision. Simulations after administrations of fixed or weight-based doses showed similar plasma concentrations. Additionally, 10 mg/kg for every other week and 15 mg/kg for every three-week administration showed comparable plasma concentrations. In conclusion, the model well described the plasma concentrations of ABL001/CTX-009 in patients with solid tumors. The simulation suggested that weight-based dose and fixed dose can provide equivalent systemic exposure.
Collapse
Affiliation(s)
- Joo Young Na
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | | | - Ki Young Huh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Sangmi Lee
- ABL Bio Inc., Seongnam, Republic of Korea
| | | | | | | | - Jaeseong Oh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Department of Pharmacology, Jeju National University College of Medicine, Jeju, Republic of Korea
- Clinical Research Institute, Jeju National University Hospital, Jeju, Republic of Korea
| |
Collapse
|
5
|
Nagaraja Shastri P, Shah N, Lechmann M, Mody H, Retter MW, Zhu M, Li T, Wang J, Shaik N, Zheng X, Ovacik M, Hua F, Jawa V, Boetsch C, Cao Y, Burke J, Datta K, Gadkar K, Upreti V, Betts A. Industry Perspective on First-in-Human and Clinical Pharmacology Strategies to Support Clinical Development of T-Cell Engaging Bispecific Antibodies for Cancer Therapy. Clin Pharmacol Ther 2024. [PMID: 39295563 DOI: 10.1002/cpt.3439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/25/2024] [Indexed: 09/21/2024]
Abstract
T-cell-engaging bispecific antibodies (TCEs) that target tumor antigens and T cells have shown great promise in treating cancer, particularly in hematological indications. The clinical development of TCEs often involves a lengthy first-in-human (FIH) trial with many dose-escalation cohorts leading up to an early proof of concept (POC), enabling either a no-go decision or dose selection for further clinical development. Multiple factors related to the target, product, disease, and patient population influence the efficacy and safety of TCEs. The intricate mechanism of action limits the translatability of preclinical models to the clinic, thereby posing challenges to streamline clinical development. In addition, unlike traditional chemotherapy, the top dose and recommended phase II doses (RP2Ds) for TCEs in the clinic are often not guided by the maximum tolerated dose (MTD), but rather based on the integrated dose-response assessment of the benefit/risk profile. These uncertainties pose complex challenges for translational and clinical pharmacologists (PK/PD scientists), as well as clinicians, to design an efficient clinical study that guides development. To that end, experts in the field, under the umbrella of the American Association of Pharmaceutical Scientists, have reviewed learnings from published literature and currently marketed products to share perspectives on the FIH and clinical pharmacology strategies to support early clinical development of TCEs.
Collapse
Affiliation(s)
- Prathap Nagaraja Shastri
- Clinical Pharmacology and Pharmacometrics, Johnson and Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | - Nirav Shah
- Clinical Pharmacology and Pharmacometrics, Johnson and Johnson Innovative Medicine, Spring House, Pennsylvania, USA
| | - Martin Lechmann
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, Penzberg, Germany
| | - Hardik Mody
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Marc W Retter
- Preclinical PK/PD, Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Min Zhu
- Clinical Pharmacology, Regeneron Pharmaceuticals, Tarrytown, New York, USA
| | - Tommy Li
- Clinical Pharmacology, Genmab, Plainsboro, New Jersey, USA
| | - Jun Wang
- Biotherapeutics Discovery Research, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, Connecticut, USA
| | - Naveed Shaik
- Clinical Pharmacology, Pfizer Oncology Development, San Diego, California, USA
| | - Xirong Zheng
- Clinical Pharmacology, Pharmacometrics and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Meric Ovacik
- Preclinical PK/PD, Genentech, South San Francisco, California, USA
| | - Fei Hua
- Certara Predictive Technology, Certara, Concord, Massachusetts, USA
| | - Vibha Jawa
- Clinical Pharmacology, Pharmacometrics and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Christophe Boetsch
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - John Burke
- Certara Predictive Technology, Certara, Concord, Massachusetts, USA
| | - Kaushik Datta
- Nonclinical Safety, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Kapil Gadkar
- Preclinical PK/PD, Genentech, South San Francisco, California, USA
| | - Vijay Upreti
- Clinical Pharmacology, Modeling & Simulation, Amgen, South San Francisco, California, USA
| | - Alison Betts
- Preclinical & Translational Sciences, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, USA
| |
Collapse
|
6
|
Tobias J, Maglakelidze M, Andrić Z, Ryspayeva D, Bulat I, Nikolić I, Petrović Z, Chawla T, Nagarkar R, Garner-Spitzer E, Zielinski CC, Chong LMO, Nixon B, Ede NJ, Yavrom S, Kundi M, Wiedermann U. Phase II Trial of HER-Vaxx, a B-cell Peptide-Based Vaccine, in HER2-Overexpressing Advanced Gastric Cancer Patients Under Platinum-Based Chemotherapy (HERIZON). Clin Cancer Res 2024; 30:4044-4054. [PMID: 39028916 PMCID: PMC11393538 DOI: 10.1158/1078-0432.ccr-24-0742] [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: 03/14/2024] [Revised: 05/01/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024]
Abstract
PURPOSE A multicenter, randomized, open-label, phase II study (HERIZON; NCT02795988) was conducted to evaluate the clinical and immunologic efficacy of HER-Vaxx (IMU-131), a B-cell, peptide-based vaccine targeting HER2 overexpressed in 6% to 30% of gastroesophageal adenocarcinomas (GEA). PATIENTS AND METHODS Patients (n = 36) with GEA were treated with standard-of-care chemotherapy (n = 17) or HER-Vaxx plus chemotherapy (n = 19), using the recommended phase 2 dose for the vaccine. Overall survival (OS; primary endpoint), safety, progression-free survival (PFS), clinical response (secondary endpoints), and vaccine-induced HER2-specific antibody levels in serum and correlation with tumor response rates (exploratory endpoints) were investigated. RESULTS A 40% OS benefit [HR, 0.60; median OS, 13.9 months; 80% confidence interval (CI), 7.52-14.32] for patients treated with HER-Vaxx plus chemotherapy compared with OS of 8.31 months (80% CI, 6.01-9.59) in patients that received chemotherapy alone. A 20% PFS difference was obtained for the vaccination arm (HR, 0.80; 80% CI, 0.47, 1.38). No additional toxicity due to HER-Vaxx was observed. The vaccine-induced high levels of HER2-specific total IgG and IgG1 antibodies (P < 0.001 vs. controls) that significantly correlated with tumor reduction (IgG, P = 0.001; IgG1, P = 0.016), had a significant capacity in inhibiting phosphorylation of the intracellular HER2-signaling pathways, mediated antibody-dependent cellular cytotoxicity, and decreased immunosuppressive FOXP3+ regulatory T cells. CONCLUSIONS HER-Vaxx plus standard chemotherapy exhibits an excellent safety profile and improves OS. Furthermore, vaccine-induced immune response was significantly associated with reduced tumor size compared with standard-of-care chemotherapy. The presented vaccination approach may substitute for treatment with trastuzumab, upon unavailability or toxicity, based on further evidence of equivalent treatment efficacy.
Collapse
Affiliation(s)
| | | | - Zoran Andrić
- Clinical Hospital Center Bezanijska Kosa, Belgrade, Serbia.
| | | | - Iurie Bulat
- ARENSIA Exploratory Medicine Research Unit, Institute of Oncology, Chisinau, Republic of Moldova.
| | - Ivan Nikolić
- Oncology Institute of Vojvodina, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia.
| | | | | | | | | | - Christoph C. Zielinski
- Central European Cancer Center, Wiener Privatklinik, Central European Cooperative Oncology Group (CECOG), Vienna, Australia.
| | | | | | | | | | | | | |
Collapse
|
7
|
Demidov L, Kharkevich G, Petenko N, Moiseenko V, Protsenko S, Semiglazova T, Zimina A, Kovalenko N, Fadeeva N, Kirtbaya D, Belogortsev I, Tantsyrev D, Odintsova S, Nesterova A, Vorontsova K, Makarycheva Y, Linkova Y, Zinkina-Orikhan A, Siliutina A, Sorokina I, Liaptseva D, Chistyakov V, Lutsky A. A phase III study to access the safety and efficacy of prolgolimab 250 mg fixed dose administered every 3 weeks versus prolgolimab 1 mg/kg every 2 weeks in patients with metastatic melanoma (FLAT). Front Oncol 2024; 14:1385685. [PMID: 39296979 PMCID: PMC11408354 DOI: 10.3389/fonc.2024.1385685] [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: 02/13/2024] [Accepted: 08/05/2024] [Indexed: 09/21/2024] Open
Abstract
Background Prolgolimab is the first Russian PD-1 inhibitor approved for the first-line treatment of unresectable or metastatic melanoma and advanced non-small cell lung cancer. It was approved in two weight-based regimens of 1 mg/kg Q2W and 3 mg/kg Q3W, but because of re-evaluation of weight-based dosing paradigm, studying of a fixed-dose regimen was considered perspective. Methods We conducted a multicenter, single-arm, open-label efficacy, pharmacokinetics, and safety study to obtain data that would allow the approval of the new flat dosing regimen of prolgolimab in patients with previously untreated unresectable or metastatic melanoma (BCD-100-8/FLAT, NCT05783882). The primary objective was to prove the non-inferiority of prolgolimab 250 mg Q3W versus prolgolimab 1 mg/kg Q2W for the treatment of patients with unresectable or metastatic melanoma in terms of ORR according to RECIST 1.1. Patients from the MIRACULUM study (BCD-100-2/MIRACULUM, NCT03269565) comprised a historical control group. Results One hundred fourteen patients received prolgolimab 250 mg Q3W, and 61 patients received prolgolimab (Prolgo) 1 mg/kg Q2W (historical control). Objective response was achieved by 33.3% [95% confidence interval (CI): 24.8, 42.8] of patients in the Prolgo 250 mg group compared with 32.8% (95% CI: 21.3, 46.0) of patients in the Prolgo 1 mg/kg group. Risk difference was 0.00, 95% CI (-0.12; NA), p = 0.0082. Both regimens were well tolerated, and safety profiles were comparable. The pharmacokinetic analysis (PK) showed that the regimen with the fixed dose of 250 mg Q3W was characterized by higher PK parameters. The immunogenicity study did not detect binding antibodies to prolgolimab in any of the subjects. Conclusion The obtained results showed that the selected fixed dosing regimen of prolgolimab 250 mg Q3W is characterized by efficacy and safety parameters comparable to that observed for the 1 mg/kg Q2W regimen.
Collapse
Affiliation(s)
- Lev Demidov
- FSBI "N.N. Blokhin National Medical Research Center of Oncology", Ministry of Health (MoH) of the Russian Federation, Moscow, Russia
| | - Galina Kharkevich
- FSBI "N.N. Blokhin National Medical Research Center of Oncology", Ministry of Health (MoH) of the Russian Federation, Moscow, Russia
| | - Natalia Petenko
- FSBI "N.N. Blokhin National Medical Research Center of Oncology", Ministry of Health (MoH) of the Russian Federation, Moscow, Russia
| | - Vladimir Moiseenko
- State Budgetary Healthcare Institution (SBHI) "St. Petersburg Clinical Scientific and Practical Center for Specialized Types of Medical Care (Oncology)", Saint Petersburg, Russia
| | - Svetlana Protsenko
- FSBI "N.N. Petrov National Medical Research Center of Oncology", Ministry of Health (MoH) of the Russian Federation, Saint Petersburg, Russia
| | - Tatiana Semiglazova
- FSBI "N.N. Petrov National Medical Research Center of Oncology", Ministry of Health (MoH) of the Russian Federation, Saint Petersburg, Russia
| | - Anastasia Zimina
- Budgetary Healthcare Institution (BHI) of the Omsk Region "Clinical Oncology Dispensary", Omsk, Russia
| | - Nadezhda Kovalenko
- State Budgetary Healthcare Institution (SBHI) "Volgograd Regional Clinical Oncology Dispensary", Volgograd, Russia
| | - Natalia Fadeeva
- State Autonomous Institution of Healthcare (SAHI) "Chelyabinsk Regional Clinical Center of Oncology and Nuclear Medicine", Chelyabinsk, Russia
| | - Dmitry Kirtbaya
- State Budgetary Healthcare Institution (SBHI) "Oncological Dispensary No. 2", Ministry of Health (MoH) of the Krasnodar Region, Krasnodar, Russia
| | - Igor Belogortsev
- Oncology Department, State Budgetary Healthcare Institution (SBHI) Leningrad Regional Clinical Hospital, Saint Petersburg, Russia
| | - Denis Tantsyrev
- Regional State Budgetary Healthcare Institution (SBHI) "Altai Regional Oncology Center", Barnaul, Russia
| | - Svetlana Odintsova
- Oncology Department, Joint-Stock Company "Modern Medical Technologies", Saint Petersburg, Russia
| | - Alfia Nesterova
- State Autonomous Institution of Healthcare (SAHI) "Professor M.Z. Sigal Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Tatarstan", Kazan, Russia
| | - Karina Vorontsova
- Moscow State Budgetary Healthcare Institution "A.S. Loginov MCSC of the Moscow City Healthcare Department", Moscow, Russia
| | - Yulia Makarycheva
- State Budgetary Healthcare Institution (SBHI) "Samara Regional Clinical Oncology Dispensary", Samara, Russia
| | - Yulia Linkova
- Clinical Research Department, Joint-Stock Company (JSC) Biocad, Saint Petersburg, Russia
| | - Arina Zinkina-Orikhan
- Clinical Research Department, Joint-Stock Company (JSC) Biocad, Saint Petersburg, Russia
| | - Anna Siliutina
- Clinical Research Department, Joint-Stock Company (JSC) Biocad, Saint Petersburg, Russia
| | - Irina Sorokina
- Moscow State Budgetary Healthcare Institution "A.S. Loginov MCSC of the Moscow City Healthcare Department", Moscow, Russia
- Oncology Department, Joint-Stock Company (JSC) Biocad, Saint Petersburg, Russia
| | - Daria Liaptseva
- Clinical Research Department, Joint-Stock Company (JSC) Biocad, Saint Petersburg, Russia
| | - Vladimir Chistyakov
- Clinical Research Department, Joint-Stock Company (JSC) Biocad, Saint Petersburg, Russia
| | - Anton Lutsky
- Clinical Research Department, Joint-Stock Company (JSC) Biocad, Saint Petersburg, Russia
| |
Collapse
|
8
|
Kicken MP, Deenen MJ, Moes DJAR, Hendrikx JJMA, van den Borne BEEM, Dumoulin DW, van der Wekken AJ, van den Heuvel MM, Ter Heine R. An Evidence-Based Rationale for Dose De-escalation of Subcutaneous Atezolizumab. Target Oncol 2024; 19:779-787. [PMID: 39085452 PMCID: PMC11393195 DOI: 10.1007/s11523-024-01087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Atezolizumab is a programmed death-ligand 1 (PD-L1) checkpoint inhibitor for the treatment of different forms of cancer. The subcutaneous formulation of atezolizumab has recently received approval. However, treatment with atezolizumab continues to be expensive, and the number of patients needing treatment with this drug continues to increase. OBJECTIVE We propose two alternative dosing regimens for subcutaneous atezolizumab to reduce drug expenses while ensuring effective exposure; one may be directly implemented in the clinic. PATIENTS AND METHODS We developed two alternative dose interval prolongation strategies based on pharmacokinetic modeling and simulation. The first dosing regimen was based on patients' weight while maintaining equivalent systemic drug exposure by adhering to Food and Drug Administration (FDA) guidelines for in silico dose adjustments. The second dosing regimen aimed to have a minimum atezolizumab concentration above the 6 µg/mL threshold, associated with 95% intratumoral PD-L1 receptor saturation for at least 95% of all patients. RESULTS We found that, for the weight-based dosing regimen, the approved 3-week dosing interval could be extended to 5 weeks for patients < 50 kg and 4 weeks for patients weighing 50-65 kg. Besides improving patient convenience, these alternative dosing intervals led to a predicted 7% and 12% cost reduction for either the USA or European population. For the second dosing regimen, we predicted that a 6-week dosing interval would result in 95% of the patients above the 6 µg/mL threshold while reducing costs by 50%. CONCLUSIONS We have developed and evaluated two alternative dosing regimens that resulted in a cost reduction. Our weight-based dosing regimen can be directly implemented and complies with FDA guidelines for alternative dosing regimens of PD-L1 inhibitors. For the more progressive alternative dosing regimen aimed at the intratumoral PD-L1 receptor threshold, further evidence on efficacy and safety is needed before implementation.
Collapse
Affiliation(s)
- Mart P Kicken
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
- Department of Clinical Pharmacy, Catharina Hospital Eindhoven, Eindhoven, The Netherlands.
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital Eindhoven, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Dirk J A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jeroen J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute (NKI-AVL), Amsterdam, The Netherlands
| | | | - Daphne W Dumoulin
- Department of Pulmonary Medicine, Erasmus Medical Center Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Anthonie J van der Wekken
- Department of Pulmonology, University of Gronigen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Arnamo AH, Pluim D, Huitema AD, Jacobs BA, Beijnen JH, Nuijen B. Prolonged in-use physicochemical and biological stability of nivolumab and pembrolizumab diluted in saline infusion bags and in partially used medication vials. J Oncol Pharm Pract 2024:10781552241279018. [PMID: 39212047 DOI: 10.1177/10781552241279018] [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: 09/04/2024]
Abstract
AIM/BACKGROUND The aim of this study was to determine long-term physicochemical and biological stability of nivolumab and pembrolizumab diluted in saline infusion bags and partially used medication vials. This may enable the prolonged clinical use of these expensive monoclonal antibodies (mAbs) to minimize the economic loss. METHODS Sterile nivolumab and pembrolizumab concentrates in partially used medication vials and compounded nivolumab and pembrolizumab infusion solutions were stored for two and four weeks, respectively, at 2-8°C in the dark. Subsequently, concentrates and compounded solutions were stored for an additional two weeks under ambient temperature and light conditions. A panel of validated and complementary methods, consisting of enzyme-linked immunosorbent assay, size exclusion chromatography, and dynamic light scattering, were used to assess the biological and physiochemical stability of these mAbs. RESULTS All samples showed that purity and concentration had remained within the criteria of <5% as stated in the European Pharmacopoeia. Diluted in infusion bags, nivolumab and pembrolizumab remained biologically and physiochemically stable for up to four weeks when stored at 2-8°C in the dark with an additional two weeks of ambient temperature and light. Stability in partially used medication vials was demonstrated for at least two weeks when stored at 2-8°C in the dark with an additional two weeks of ambient temperature and light. CONCLUSION The findings of this study justify the storage and clinical re-use of sterile nivolumab and pembrolizumab in partially used medication vials and compounded IV infusion bags for up to six weeks. This minimizes the risk of economic loss due to waste. Moreover, these findings support the batch-wise compounding of fixed-dose and dose-banded nivolumab and pembrolizumab infusion bags.
Collapse
Affiliation(s)
- A Hedvig Arnamo
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Dick Pluim
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alwin Dr Huitema
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Bart Aw Jacobs
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, Antoni van Leeuwenhoek hospital - Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Li L, Pang J, Yan Y, Zhang Q, Zheng S, Chen M, Yi W, Wu J. Comparing long-term prognosis following different surgical methods in patients with early stage breast cancer and obesity: a retrospective cohort study in China. BMJ Open 2024; 14:e078816. [PMID: 39214662 PMCID: PMC11367398 DOI: 10.1136/bmjopen-2023-078816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Breast-conserving therapy (BCT) includes breast-conserving surgery (BCS) combined with radiation therapy (RT). RT plays a crucial role in improving the prognosis of patients who undergo BCS. However, obesity is a potential risk factor for resistance to radiation. The aim of this study was to evaluate any difference in the long-term prognosis of patients with early stage breast cancer and obesity treated with BCT or total mastectomy (TM). DESIGN, SETTING AND PARTICIPANTS This was a retrospective cohort study involving 1125 patients diagnosed with early stage breast cancer and obesity at the Shanghai Cancer Center of Fudan University from 2013 to 2016. OUTCOME MEASURES Obesity in the Chinese population was defined as a body mass index ≥28 kg/m2. Surgical options included BCT and TM. The primary survival outcomes were overall survival (OS), disease-free survival (DFS) and recurrence-free survival (RFS). Inverse probability of treatment weighting (IPTW) was used to control for the impact of confounding factors on prognosis. RESULTS The median follow-up times in the BCT group and TM group without postoperative RT were 51.1 months (IQR of 40.6-68.1 months) and 61.8 months (IQR of 46.5-76.7 months), respectively. After IPTW, the baseline data were balanced. Compared with those in the TM cohort, patients in the whole IPTW cohort in the BCT cohort had worse DFS (HR 4.280, 95% CI 2.180 to 8.400; p<0.001), RFS (HR 4.380, 95% CI 2.370 to 8.120; p<0.001) and OS (HR 3.590, 95% CI 1.620 to 7.950; p=0.002). CONCLUSION In patients with early stage breast cancer and obesity, TM is associated with better survival outcomes than BCT.
Collapse
Affiliation(s)
- Lun Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, China
| | - Jian Pang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, China
| | - Yiqing Yan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, China
| | - Qi Zhang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shuyue Zheng
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ming Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Research Center for Breast Disease in Hunan Province, Changsha, China
| | - Jiong Wu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Collaborative Innovation Center for Cancer Medicine, Shanghai, China
| |
Collapse
|
11
|
Senaldi G, Mohan A, Zhang L, Tanaka J, Lin Y, Pandya G, Grossman S, Urbina S, Reynolds SH, Hand AH. First-in-Human Study of the Safety, Tolerability, Pharmacokinetics, Immunogenicity, and Pharmacodynamics of DS-7011a, an Anti-TLR7 Antagonistic Monoclonal Antibody for the Treatment of Systemic Lupus Erythematosus. J Clin Pharmacol 2024. [PMID: 39169827 DOI: 10.1002/jcph.6117] [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: 04/25/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Toll-like receptor (TLR)7 is a pattern recognition receptor that critically contributes to the pathogenesis of systemic lupus erythematosus (SLE). DS-7011a is an anti-TLR7 monoclonal antibody that prevents TLR7 from signaling. The aim of this first-in-human, double-blind, randomized, and placebo-controlled study was to evaluate the safety, pharmacokinetics, immunogenicity, and pharmacodynamics of single ascending intravenous (IV) and subcutaneous (SC) doses of DS-7011a in healthy subjects (HS) (NCT05203692). On day 1, 80 HS received DS-7011a or placebo 6:2 in 10 cohorts (7 treated IV and 3 SC) of 8 each and were followed for 8 weeks until day 57. Safety was evaluated by recording treatment-emergent adverse events (TEAEs), pharmacokinetics by measuring plasma DS-7011a, immunogenicity by measuring plasma anti-drug antibodies (ADAs), and pharmacodynamics by evaluating the suppression of interleukin-6 production ex vivo in whole blood. DS-7011a was safe and well tolerated across all cohorts. TEAEs were mostly mild in severity and not drug-related. DS-7011a exposure increased with the dose but was not dose proportional, as the elimination of lower doses was accelerated by target-mediated drug disposition. Terminal half-life was about 15-17 days and Tmax upon SC administration was about 5 days. DS-7011a induced ADAs in about half of HS but with no impact on clinical findings and pharmacokinetics. Pharmacodynamic (PD) response also increased with the dose and at the higher doses was of large extent (>90%), early onset, and lasting duration. DS-7011a showed favorable safety, pharmacokinetics, immunogenicity, and PD properties that support its development for the treatment of SLE.
Collapse
Affiliation(s)
| | | | - Li Zhang
- Daiichi Sankyo, Basking Ridge, NJ, USA
| | | | - Yong Lin
- Daiichi Sankyo, Basking Ridge, NJ, USA
| | | | | | | | | | - Alan H Hand
- Worldwide Clinical Trials, San Antonio, TX, USA
| |
Collapse
|
12
|
Stalder T, Andre C, Ben Mahi M, Lethier L, Limat S, Legat C, Guillaume Y. Performance evaluation of the HPLC diode array and evaporative light scattering detection for the implementation of dose-banded gemcitabine infusion bags. Eur J Hosp Pharm 2024; 31:369-375. [PMID: 36737226 PMCID: PMC11265545 DOI: 10.1136/ejhpharm-2022-003540] [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: 09/01/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Dose banding (DB) was used to optimise the individualisation of patient treatments with gemcitabine (Gem) in order to improve workload planning at the pharmacy of the University Hospital Centre of Besançon (UHCB). A new simple and fast high-performance liquid chromatographic (HPLC) method was also developed for the quantification of Gem without dilution of the infusion bags. METHODS Individual doses of Gem preparations were retrospectively analysed over a 1-year period to determine the frequency of prepared doses. Using a maximum gap of 7.5% around the doses chosen, the selected Gem standard doses were 1400 mg, 1600 mg, 1800 mg and 2000 mg. Following the DB scheme, the frequency of prescription of standard and individualised Gem doses was analysed over a period of 10 months. The four selected Gem standard doses were aseptically prepared in polyolefin infusion bags. Each series of 20 bags was stocked under refrigerated storage conditions (4°C) for up to 84 days. The quantification of Gem without dilution of the infusion bags was obtained by the development of a HPLC method coupled to a diode array detector (DAD) or an evaporative light scattering detector (ELSD). RESULTS During the 10-month period following implementation of the DB, 75.6% of the 1266 prescribed doses were covered by the four standardised preparations. The number of different Gem doses was reduced from 183 to 55. Concerning the Gem quantification, both heteroscedasticity and non-linearity were observed with DAD. Using an ELSD, the trueness values were between 98.59% and 101.52% with excellent repeatability values between 0.66% and 1.42%. CONCLUSION A new HPLC method has been developed for the quantification of Gem without dilution of the infusion bags prepared in advance as a result of a target DB scheme successfully implemented in our pharmacy department.
Collapse
MESH Headings
- Deoxycytidine/analysis
- Deoxycytidine/analogs & derivatives
- Chromatography, High Pressure Liquid/methods
- Chromatography, High Pressure Liquid/standards
- Gemcitabine
- Antimetabolites, Antineoplastic/analysis
- Antimetabolites, Antineoplastic/administration & dosage
- Antimetabolites, Antineoplastic/standards
- Drug Packaging/standards
- Drug Packaging/methods
- Infusions, Intravenous/methods
- Infusions, Intravenous/standards
- Infusions, Intravenous/instrumentation
- Retrospective Studies
- Humans
- Scattering, Radiation
- Light
- Pharmacy Service, Hospital/methods
- Pharmacy Service, Hospital/standards
Collapse
Affiliation(s)
- Thomas Stalder
- Franche-Comte University Faculty of Medical and Pharmaceutical Sciences, Besançon, France
| | - Claire Andre
- Franche-Comte University Faculty of Medical and Pharmaceutical Sciences, Besançon, France
| | | | - Lydie Lethier
- Franche-Comte University Faculty of Medical and Pharmaceutical Sciences, Besançon, France
| | | | | | - Yves Guillaume
- Franche-Comte University Faculty of Medical and Pharmaceutical Sciences, Besançon, France
| |
Collapse
|
13
|
Van Cauwenberge J, Van Baelen K, Maetens M, Geukens T, Nguyen HL, Nevelsteen I, Smeets A, Deblander A, Neven P, Koolen S, Wildiers H, Punie K, Desmedt C. Reporting on patient's body mass index (BMI) in recent clinical trials for patients with breast cancer: a systematic review. Breast Cancer Res 2024; 26:81. [PMID: 38778365 PMCID: PMC11112918 DOI: 10.1186/s13058-024-01832-7] [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: 11/27/2023] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The proportion of patients with breast cancer and obesity is increasing. While the therapeutic landscape of breast cancer has been expanding, we lack knowledge about the potential differential efficacy of most drugs according to the body mass index (BMI). Here, we conducted a systematic review on recent clinical drug trials to document the dosing regimen of recent drugs, the reporting of BMI and the possible exclusion of patients according to BMI, other adiposity measurements and/or diabetes (leading comorbidity of obesity). We further explored whether treatment efficacy was evaluated according to BMI. METHODS A search of Pubmed and ClinicalTrials.gov was performed to identify phase I-IV trials investigating novel systemic breast cancer treatments. Dosing regimens and exclusion based on BMI, adiposity measurements or diabetes, documentation of BMI and subgroup analyses according to BMI were assessed. RESULTS 495 trials evaluating 26 different drugs were included. Most of the drugs (21/26, 81%) were given in a fixed dose independent of patient weight. BMI was an exclusion criterion in 3 out of 495 trials. Patients with diabetes, the leading comorbidity of obesity, were excluded in 67/495 trials (13.5%). Distribution of patients according to BMI was mentioned in 8% of the manuscripts, subgroup analysis was performed in 2 trials. No other measures of adiposity/body composition were mentioned in any of the trials. Retrospective analyses on the impact of BMI were performed in 6 trials. CONCLUSIONS Patient adiposity is hardly considered as most novel drug treatments are given in a fixed dose. BMI is generally not reported in recent trials and few secondary analyses are performed. Given the prevalence of patients with obesity and the impact obesity can have on pharmacokinetics and cancer biology, more attention should be given by investigators and study sponsors to reporting patient's BMI and evaluating its impact on treatment efficacy and toxicity.
Collapse
Affiliation(s)
- Josephine Van Cauwenberge
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Karen Van Baelen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
| | - Tatjana Geukens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Ha Linh Nguyen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium
| | - Ines Nevelsteen
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Ann Smeets
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Anne Deblander
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Stijn Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC, Rotterdam, The Netherlands
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Kevin Punie
- Department of Medical Oncology, GZA Hospitals Sint-Augustinus, Wilrijk, Belgium
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Herestraat 49, Box 808, 3000, Louvain, Belgium.
| |
Collapse
|
14
|
Taylor KS, McMonagle MM, Guy SC, Human-McKinnon AM, Asamizu S, Fletcher HJ, Davis BW, Suyama TL. Albumin-ruthenium catalyst conjugate for bio-orthogonal uncaging of alloc group. Org Biomol Chem 2024; 22:2992-3000. [PMID: 38526322 DOI: 10.1039/d4ob00234b] [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] [Indexed: 03/26/2024]
Abstract
The employment of antibodies as a targeted drug delivery vehicle has proven successful which is exemplified by the emergence of antibody-drug conjugates (ADCs). However, ADCs are not without their shortcomings. Improvements may be made to the ADC platform by decoupling the cytotoxic drug from the delivery vehicle and conjugating an organometallic catalyst in its place. The resulting protein-metal catalyst conjugate was designed to uncage the masked cytotoxin administered as a separate entity. Macropinocytosis of albumin by cancerous cells suggests the potential of albumin acting as the tumor-targeting delivery vehicle. Herein reported are the first preparation and demonstration of ruthenium catalysts with cyclopentadienyl and quinoline-based ligands conjugated to albumin. The effective uncaging abilities were demonstrated on allyloxy carbamate (alloc)-protected rhodamine 110 and doxorubicin, providing a promising catalytic scaffold for the advancement of selective drug delivery methods in the future.
Collapse
Affiliation(s)
- Kimberly S Taylor
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Madison M McMonagle
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Schaelee C Guy
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Ariana M Human-McKinnon
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Shumpei Asamizu
- Engineering Biology Research Center, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Heidi J Fletcher
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Bradley W Davis
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Takashi L Suyama
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| |
Collapse
|
15
|
Pluchart H, Chanoine S, Moro-Sibilot D, Chouaid C, Frey G, Villa J, Degano B, Giaj Levra M, Bedouch P, Toffart AC. Lung cancer, comorbidities, and medication: the infernal trio. Front Pharmacol 2024; 14:1016976. [PMID: 38450055 PMCID: PMC10916800 DOI: 10.3389/fphar.2023.1016976] [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: 08/11/2022] [Accepted: 09/25/2023] [Indexed: 03/08/2024] Open
Abstract
Most patients with lung cancer are smokers and are of advanced age. They are therefore at high risk of having age- and lifestyle-related comorbidities. These comorbidities are subject to treatment or even polypharmacy. There is growing evidence of a link between lung cancer, comorbidities and medications. The relationships between these entities are complex. The presence of comorbidities and their treatments influence the time of cancer diagnosis, as well as the diagnostic and treatment strategy. On the other hand, cancer treatment may have an impact on the patient's comorbidities such as renal failure, pneumonitis or endocrinopathies. This review highlights how some comorbidities may have an impact on lung cancer presentation and may require treatment adjustments. Reciprocal influences between the treatment of comorbidities and anticancer therapy will also be discussed.
Collapse
Affiliation(s)
- Hélène Pluchart
- Pôle Pharmacie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, France
- Université Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, CNRS, Grenoble INP, TIMC UMR5525, Grenoble, France
| | - Sébastien Chanoine
- Pôle Pharmacie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, France
- Université Grenoble Alpes, Grenoble, France
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
| | - Denis Moro-Sibilot
- Université Grenoble Alpes, Grenoble, France
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Christos Chouaid
- Service de Pneumologie, Centre Hospitalier Intercommunal de Créteil, Créteil, France
- Inserm U955, UPEC, IMRB, équipe CEpiA, CréteilFrance
| | - Gil Frey
- Service de Chirurgie Thoracique, Vasculaire et Endocrinienne, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Julie Villa
- Service de Radiothérapie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Bruno Degano
- Université Grenoble Alpes, Grenoble, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
- Laboratoire HP2, INSERM U1042, Université Grenoble Alpes, Grenoble, France
| | - Matteo Giaj Levra
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Pierrick Bedouch
- Pôle Pharmacie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, France
- Université Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, CNRS, Grenoble INP, TIMC UMR5525, Grenoble, France
| | - Anne-Claire Toffart
- Université Grenoble Alpes, Grenoble, France
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| |
Collapse
|
16
|
Reinig S, Shih SR. Non-neutralizing functions in anti-SARS-CoV-2 IgG antibodies. Biomed J 2024; 47:100666. [PMID: 37778697 PMCID: PMC10825350 DOI: 10.1016/j.bj.2023.100666] [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: 07/01/2023] [Revised: 08/31/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023] Open
Abstract
Most individuals infected with or vaccinated against COVID-19 develop antigenic neutralizing immunoglobulin G (IgG) antibodies against the SARS-CoV-2 spike protein. Although neutralizing antibodies are biomarkers of the adaptive immune response, their mere presence is insufficient to explain the protection afforded against the disease or its pathology. IgG exhibits other secondary effector functions that activate innate immune components, including complement, natural killer cells, and macrophages. The affinity for effector cells depends on the isotypes and glycosylation of IgG antibodies. The anti-spike IgG titer should be sufficient to provide significant Fc-mediated effects in severe COVID-19, mRNA, and protein subunit vaccinations. In combination with aberrant effector cells, pro-inflammatory afucosylated IgG1 and IgG3 may be detrimental in severe COVID-19. The antibody response of mRNA vaccines leads to higher fucosylation and a less inflammatory IgG profile, with a long-term shift to IgG4, which is correlated with protection from disease.
Collapse
Affiliation(s)
- Sebastian Reinig
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
17
|
Roy S, Pan Z, Abu Qarnayn N, Alajmi M, Alatawi A, Alghamdi A, Alshaoosh I, Asiri Z, Batista B, Chaturvedi S, Dehinsilu O, Edduweh H, El-Adawy R, Hossen E, Mojra B, Rana J. A robust optimal control framework for controlling aberrant RTK signaling pathways in esophageal cancer. J Math Biol 2024; 88:14. [PMID: 38180543 DOI: 10.1007/s00285-023-02033-0] [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: 04/02/2023] [Revised: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024]
Abstract
This study presents a new framework for obtaining personalized optimal treatment strategies targeting aberrant signaling pathways in esophageal cancer, such as the epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF) signaling pathways. A new pharmacokinetic model is developed taking into account specific heterogeneities of these signaling mechanisms. The optimal therapies are designed to be obtained using a three step process. First, a finite-dimensional constrained optimization problem is solved to obtain the parameters of the pharmacokinetic model, using discrete patient data measurements. Next, a sensitivity analysis is carried out to determine which of the parameters are sensitive to the evolution of the variants of EGF receptors and VEGF receptors. Finally, a second optimal control problem is solved based on the sensitivity analysis results, using a modified pharmacokinetic model that incorporates two representative drugs Trastuzumab and Bevacizumab, targeting EGF and VEGF, respectively. Numerical results with the combination of the two drugs demonstrate the efficiency of the proposed framework.
Collapse
Affiliation(s)
- Souvik Roy
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA.
| | - Zui Pan
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Naif Abu Qarnayn
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Mesfer Alajmi
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Ali Alatawi
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Asma Alghamdi
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Ibrahem Alshaoosh
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Zahra Asiri
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Berlinda Batista
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Shreshtha Chaturvedi
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Olusola Dehinsilu
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Hussein Edduweh
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Rodina El-Adawy
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Emran Hossen
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Bardia Mojra
- Department of Computer Science, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| | - Jashmon Rana
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, 76019-0407, USA
| |
Collapse
|
18
|
Erstad BL, Davis LE. Fixed Versus Body-Sized-Based Dosing of Monoclonal Antibodies. Ann Pharmacother 2024; 58:91-95. [PMID: 37129243 DOI: 10.1177/10600280231170650] [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] [Indexed: 05/03/2023] Open
Abstract
Monoclonal antibody products are an increasing portion of novel drug approvals. The labeling of initial drug approvals frequently involves body-size-based rather than fixed-dose administration regimens for adults without clear rationale for doing so. This presents challenges when prescribing these products for patients with extremes of body habitus who constitute a small portion of enrollment in pre-approval investigations. Fixed-dose regimens allow for standardized preparation with the potential to reduce the risk of calculation errors, drug waste, and make home administration more practical. Fixed-dose rather than body-size-based monoclonal antibody regimens should serve as the initial approach in early phase 1 clinical trials.
Collapse
Affiliation(s)
- Brian L Erstad
- Department of Pharmacy Practice & Science, The University of Arizona College of Pharmacy, Tucson, AZ, USA
| | - Lisa E Davis
- Department of Pharmacy Practice & Science, The University of Arizona College of Pharmacy, Tucson, AZ, USA
| |
Collapse
|
19
|
Dumoulin DW, Douma LH, Hofman MM, van der Noort V, Cornelissen R, de Gooijer CJ, Burgers JA, Aerts JGJV. Nivolumab and ipilimumab in the real-world setting in patients with mesothelioma. Lung Cancer 2024; 187:107440. [PMID: 38104353 DOI: 10.1016/j.lungcan.2023.107440] [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: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVES Nivolumab (anti-PD-1) plus ipilimumab (anti-CTLA-4) is a new first-line treatment combination for patients with pleural mesothelioma. Nivolumab-ipilimumab improved the survival, however, 30.3% of the patients suffered from grade 3-4 treatment related adverse events (TRAE's) and TRAE's led to discontinuation in 23.0% of all patients. Here, we present the first real-world data of nivolumab plus ipilimumab in patients with malignant mesothelioma treated in two mesothelioma expert centers. METHODS Clinical data of patients with mesothelioma treated with nivolumab and ipilimumab were prospectively collected. Clinical parameters were obtained every visit, CT scans were evaluated every 12 weeks and adverse events were assessed continuously during the treatment. Data on grade 2-5 TRAE's and activity (overall response rate (ORR), duration of response (DOR), disease control rate (DCR), median progression-free survival (mPFS) and median overall survival (mOS) were reported. RESULTS Between January 2021 and August 2022, 184 patients were treated with nivolumab plus ipilimumab. The median follow-up was 12.1 months (95 %CI 11.1 - 13.1). Grade 3-4 TRAEs were seen in 27.7 % of the patients and 25.0 % discontinued immunotherapy treatment early because of TRAE's. ORR was 21.7 % (95 % CI 15.7-27.7), median DOR was 5.7 months (IQR 3.2-8.7) and DCR at 12 weeks 56.0 % (95 % CI 48.8-63.2). The mPFS was 5.5 months (95 %CI 4.1-6.9), mOS was 14.1 months (95 % CI 11.1-18.2). CONCLUSIONS Nivolumab plus ipilimumab had an equal efficacy in a real-world comparable population but also a high risk of TRAE's, leading to discontinuation of treatment in 25% of the patients.
Collapse
Affiliation(s)
- D W Dumoulin
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands.
| | - L H Douma
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M M Hofman
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands; Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands
| | - V van der Noort
- Department of Biometrics, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - R Cornelissen
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands
| | - C J de Gooijer
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J A Burgers
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands
| |
Collapse
|
20
|
Desai A, Xiao A, Smith C, Jensen C, Pritchett J, Soefje S, Durani U, Go R, Scheckel C. Analysis of Medicare Expenditure for Discarded Infused Cancer Therapeutics From 2017-2020. Mayo Clin Proc 2023; 98:1767-1773. [PMID: 38043994 DOI: 10.1016/j.mayocp.2023.07.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/26/2023] [Accepted: 07/19/2023] [Indexed: 12/05/2023]
Abstract
OBJECTIVE To explore patterns in Medicare reimbursement for wasted oncologic and hematologic infusion drugs from 2017 to 2020 and estimate the savings that implementation of the Infrastructure Investment and Jobs Act (IIJA) would have had. METHODS Using the publicly available Medicare Part B Discarded Drug Units database, we analyzed reimbursement data for discarded antineoplastic and hematology therapies from 2017 to 2020. RESULTS Medicare Part B utilization data was extracted for 77 therapies. From 2017 to 2020, the median annual dollar value of discarded therapies was $590 million. Every year, bortezomib, azacitidine, cabazitaxel, and decitabine were among the most wasted products, an average 24% waste. The IIJA policy would have impacted a median of 20 oncology agents and resulted in median annual refund of $172 million. Had the top five most discarded therapies been redistributed, they could have treated 18,289 patients. The five most wasted drugs were all dosed by weight and distributed in single-use vials. CONCLUSION The IIJA could potentially significantly reduce waste or encourage redistribution to treat thousands of additional patients. We propose that a fusion of fixed and weight-based dosing may help reduce wasteful medication administration by offering doses that better accommodate most patients. We anticipate that manufacturers will adapt to the IIJA perhaps by adjusting fixed doses or simply increasing drug prices. If price changes from dose delivery adjustment occur, rebates offered to pharmacy benefit managers and insurers will likely follow suit and may alter formulary positioning.
Collapse
Affiliation(s)
- Aakash Desai
- Division of Hematology, Mayo Clinic, Rochester, MN, USA; Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Alexander Xiao
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Caleb Smith
- Division of Hematology, Mayo Clinic, Rochester, MN, USA; Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Chelsee Jensen
- Division of Supply Chain Management, Mayo Clinic, Rochester, MN, USA
| | - Joshua Pritchett
- Division of Hematology, Mayo Clinic, Rochester, MN, USA; Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Scott Soefje
- Division of Hematology, Mayo Clinic, Rochester, MN, USA; Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA; Department of Pharmacy, Mayo Clinic, Rochester, MN, USA
| | | | - Ronald Go
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Caleb Scheckel
- Division of Hematology, Mayo Clinic, Rochester, MN, USA; Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
21
|
Cao C, Ligibel JA. Too Much, Too Little, or Just Right? Obesity and Dosing of Targeted Therapies in Breast Cancer. J Clin Oncol 2023; 41:5090-5092. [PMID: 37797274 PMCID: PMC10666978 DOI: 10.1200/jco.23.01516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/10/2023] [Indexed: 10/07/2023] Open
Affiliation(s)
- Chao Cao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | |
Collapse
|
22
|
Meza NP, Hardy CA, Morin KH, Huang C, Raghava S, Song J, Zhang J, Wang Y. Predicting Colloidal Stability of High-Concentration Monoclonal Antibody Formulations in Common Pharmaceutical Buffers Using Improved Polyethylene Glycol Induced Protein Precipitation Assay. Mol Pharm 2023; 20:5842-5855. [PMID: 37867303 DOI: 10.1021/acs.molpharmaceut.3c00694] [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] [Indexed: 10/24/2023]
Abstract
Colloidal stability is an important consideration when developing high concentration mAb formulations. PEG-induced protein precipitation is a commonly used assay to assess the colloidal stability of protein solutions. However, the practical usefulness and the current theoretical model for this assay have yet to be verified over a large formulation space across multiple mAbs and mAb-based modalities. In the present study, we used PEG-induced protein precipitation assays to evaluate colloidal stability of 3 mAbs in 24 common formulation buffers at 20 and 5 °C. These prediction assays were conducted at low protein concentration (1 mg/mL). We also directly characterized high concentration (100 mg/mL) formulations for cold-induced phase separation, turbidity, and concentratibility by ultrafiltration. This systematic study allowed analysis of the correlation between the results of low concentration assays and the high concentration attributes. The key findings of this study include the following: (1) verification of the usefulness of three different parameters (Cmid, μB, and Tcloud) from PEG-induced protein precipitation assays for ranking colloidal stability of high concentration mAb formulations; (2) a new method to implement PEG-induced protein precipitation assay suitable for high throughput screening with low sample consumption; (3) improvement in the theoretical model for calculating robust thermodynamic parameters of colloidal stability (μB and εB) that are independent of specific experimental settings; (4) systematic evaluation of the effects of pH and buffer salts on colloidal stability of mAbs in common formulation buffers. These findings provide improved theoretical and practical tools for assessing the colloidal stability of mAbs and mAb-based modalities during formulation development.
Collapse
Affiliation(s)
- Noemi P Meza
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Colin A Hardy
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Kylie H Morin
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Chengbin Huang
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Smita Raghava
- Sterile and Specialty Products, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jing Song
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jingtao Zhang
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ying Wang
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| |
Collapse
|
23
|
Zamboni WC, Charlab R, Burckart GJ, Stewart CF. Effect of Obesity on the Pharmacokinetics and Pharmacodynamics of Anticancer Agents. J Clin Pharmacol 2023; 63 Suppl 2:S85-S102. [PMID: 37942904 DOI: 10.1002/jcph.2326] [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: 05/01/2023] [Accepted: 07/12/2023] [Indexed: 11/10/2023]
Abstract
An objective of the Precision Medicine Initiative, launched in 2015 by the US Food and Drug Administration and National Institutes of Health, is to optimize and individualize dosing of drugs, especially anticancer agents, with high pharmacokinetic and pharmacodynamic variability. The American Society of Clinical Oncology recently reported that 40% of obese patients receive insufficient chemotherapy doses and exposures, which may lead to reduced efficacy, and recommended pharmacokinetic studies to guide appropriate dosing in these patients. These issues will only increase in importance as the incidence of obesity in the population increases. This publication reviews the effects of obesity on (1) tumor biology, development of cancer, and antitumor response; (2) pharmacokinetics and pharmacodynamics of small-molecule anticancer drugs; and (3) pharmacokinetics and pharmacodynamics of complex anticancer drugs, such as carrier-mediated agents and biologics. These topics are not only important from a scientific research perspective but also from a drug development and regulator perspective. Thus, it is important to evaluate the effects of obesity on the pharmacokinetics and pharmacodynamics of anticancer agents in all categories of body habitus and especially in patients who are obese and morbidly obese. As the effects of obesity on the pharmacokinetics and pharmacodynamics of anticancer agents may be highly variable across drug types, the optimal dosing metric and algorithm for difference classes of drugs may be widely different. Thus, studies are needed to evaluate current and novel metrics and methods for measuring body habitus as related to optimizing the dose and reducing pharmacokinetic and pharmacodynamic variability of anticancer agents in patients who are obese and morbidly obese.
Collapse
Affiliation(s)
- William C Zamboni
- UNC Eshelman School of Pharmacy, UNC Lineberger Comprehensive Cancer Center, Caroline Institute of Nanomedicine, University of North Carolina, Chapel Hill, NC, USA
| | - Rosane Charlab
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Gilbert J Burckart
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | | |
Collapse
|
24
|
Buyel JF. Product safety aspects of plant molecular farming. Front Bioeng Biotechnol 2023; 11:1238917. [PMID: 37614627 PMCID: PMC10442644 DOI: 10.3389/fbioe.2023.1238917] [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: 06/12/2023] [Accepted: 07/31/2023] [Indexed: 08/25/2023] Open
Abstract
Plant molecular farming (PMF) has been promoted since the 1990s as a rapid, cost-effective and (most of all) safe alternative to the cultivation of bacteria or animal cells for the production of biopharmaceutical proteins. Numerous plant species have been investigated for the production of a broad range of protein-based drug candidates. The inherent safety of these products is frequently highlighted as an advantage of PMF because plant viruses do not replicate in humans and vice versa. However, a more nuanced analysis of this principle is required when considering other pathogens because toxic compounds pose a risk even in the absence of replication. Similarly, it is necessary to assess the risks associated with the host system (e.g., the presence of toxic secondary metabolites) and the production approach (e.g., transient expression based on bacterial infiltration substantially increases the endotoxin load). This review considers the most relevant host systems in terms of their toxicity profile, including the presence of secondary metabolites, and the risks arising from the persistence of these substances after downstream processing and product purification. Similarly, we discuss a range of plant pathogens and disease vectors that can influence product safety, for example, due to the release of toxins. The ability of downstream unit operations to remove contaminants and process-related toxic impurities such as endotoxins is also addressed. This overview of plant-based production, focusing on product safety aspects, provides recommendations that will allow stakeholders to choose the most appropriate strategies for process development.
Collapse
Affiliation(s)
- J. F. Buyel
- Department of Biotechnology (DBT), Institute of Bioprocess Science and Engineering (IBSE), University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| |
Collapse
|
25
|
Bittner B, Sánchez-Félix M, Lee D, Koynov A, Horvath J, Schumacher F, Matoori S. Drug delivery breakthrough technologies - A perspective on clinical and societal impact. J Control Release 2023; 360:335-343. [PMID: 37364797 DOI: 10.1016/j.jconrel.2023.06.034] [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: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
The way a drug molecule is administered has always had a profound impact on people requiring medical interventions - from vaccine development to cancer therapeutics. In the Controlled Release Society Fall Symposium 2022, a trans-institutional group of scientists from industry, academia, and non-governmental organizations discussed what a breakthrough in the field of drug delivery constitutes. On the basis of these discussions, we classified drug delivery breakthrough technologies into three categories. In category 1, drug delivery systems enable treatment for new molecular entities per se, for instance by overcoming biological barriers. In category 2, drug delivery systems optimize efficacy and/or safety of an existing drug, for instance by directing distribution to their target tissue, by replacing toxic excipients, or by changing the dosing reqimen. In category 3, drug delivery systems improve global access by fostering use in low-resource settings, for instance by facilitating drug administration outside of a controlled health care institutional setting. We recognize that certain breakthroughs can be classified in more than one category. It was concluded that in order to create a true breakthrough technology, multidisciplinary collaboration is mandated to move from pure technical inventions to true innovations addressing key current and emerging unmet health care needs.
Collapse
Affiliation(s)
- Beate Bittner
- Global Product Strategy, Product Optimization, Grenzacher Strasse 124, 4070 Basel, Switzerland.
| | - Manuel Sánchez-Félix
- Novartis Institutes for BioMedical Research, 700 Main Street, Cambridge, MA 02139, USA
| | - Dennis Lee
- Bill & Melinda Gates Foundation, Seattle, WA 98119, United States
| | - Athanas Koynov
- Pharmaceutical Sciences, Merck & Co., Inc., Rahway, NJ 07033, United States
| | - Joshua Horvath
- Device and Packaging Development, Genentech, Inc., South San Francisco, CA, United States
| | - Felix Schumacher
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - Simon Matoori
- Faculté de Pharmacie, Université de Montréal, 2940 Chemin de Polytechnique, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
26
|
Mansour I, Murugapandian S, Tanriover B, Thajudeen B. Contemporary Monoclonal Antibody Utilization in Glomerular Diseases. Mayo Clin Proc Innov Qual Outcomes 2023; 7:276-290. [PMID: 37448529 PMCID: PMC10338194 DOI: 10.1016/j.mayocpiqo.2023.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023] Open
Abstract
Therapeutic monoclonal antibodies (MAbs) have been one of the fastest growing drug classes in the past 2 decades and are indicated in the treatment of cancer, autoimmune disorders, solid organ transplantation, and glomerular diseases. The Food and Drug Administration has approved 100 MAbs between 1986 and 2021, and MAbs account for 20% of Food and Drug Administration's new drug approval every year. MAbs are preferred over traditional immunosuppressive agents because of their high specificity, reduced number of drug-drug interactions, and low toxicity, which make them a prime example of personalized medicine. In this review article, we provide an overview of the taxonomy, pharmacology, and therapeutic applications of MAbs in glomerular diseases. We searched the literature through PubMed using the following search terms: monoclonal antibodies, glomerular diseases, pharmacokinetics, pharmacodynamics, immunoglobulin, murine, chimeric,humanized, and fully human, and limited our search to years 2018-2023. We selected peer-reviewed journal articles with an evidence-based approach, prioritizing randomized control trials in specific glomerular diseases, if available. Advances in the MAb field have resulted in a significant paradigm shift in targeted treatment of immune-mediated glomerular diseases, and multiple randomized control trials are currently being conducted. Increased recognition is critical to expand their use in experimental research and personalized medicine.
Collapse
Affiliation(s)
- Iyad Mansour
- Division of Nephrology, College of Medicine, The University of Arizona, Tucson
| | | | - Bekir Tanriover
- Division of Nephrology, College of Medicine, The University of Arizona, Tucson
| | - Bijin Thajudeen
- Division of Nephrology, College of Medicine, The University of Arizona, Tucson
| |
Collapse
|
27
|
Seaton KE, Huang Y, Karuna S, Heptinstall JR, Brackett C, Chiong K, Zhang L, Yates NL, Sampson M, Rudnicki E, Juraska M, deCamp AC, Edlefsen PT, Mullins JI, Williamson C, Rossenkhan R, Giorgi EE, Kenny A, Angier H, Randhawa A, Weiner JA, Rojas M, Sarzotti-Kelsoe M, Zhang L, Sawant S, Ackerman ME, McDermott AB, Mascola JR, Hural J, McElrath MJ, Andrew P, Hidalgo JA, Clark J, Laher F, Orrell C, Frank I, Gonzales P, Edupuganti S, Mgodi N, Corey L, Morris L, Montefiori D, Cohen MS, Gilbert PB, Tomaras GD. Pharmacokinetic serum concentrations of VRC01 correlate with prevention of HIV-1 acquisition. EBioMedicine 2023; 93:104590. [PMID: 37300931 PMCID: PMC10363420 DOI: 10.1016/j.ebiom.2023.104590] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND The phase 2b proof-of-concept Antibody Mediated Prevention (AMP) trials showed that VRC01, an anti-HIV-1 broadly neutralising antibody (bnAb), prevented acquisition of HIV-1 sensitive to VRC01. To inform future study design and dosing regimen selection of candidate bnAbs, we investigated the association of VRC01 serum concentration with HIV-1 acquisition using AMP trial data. METHODS The case-control sample included 107 VRC01 recipients who acquired HIV-1 and 82 VRC01 recipients who remained without HIV-1 during the study. We measured VRC01 serum concentrations with a qualified pharmacokinetic (PK) Binding Antibody Multiplex Assay. We employed nonlinear mixed effects PK modelling to estimate daily-grid VRC01 concentrations. Cox regression models were used to assess the association of VRC01 concentration at exposure and baseline body weight, with the hazard of HIV-1 acquisition and prevention efficacy as a function of VRC01 concentration. We also compared fixed dosing vs. body weight-based dosing via simulations. FINDINGS Estimated VRC01 concentrations in VRC01 recipients without HIV-1 were higher than those in VRC01 recipients who acquired HIV-1. Body weight was inversely associated with HIV-1 acquisition among both placebo and VRC01 recipients but did not modify the prevention efficacy of VRC01. VRC01 concentration was inversely correlated with HIV-1 acquisition, and positively correlated with prevention efficacy of VRC01. Simulation studies suggest that fixed dosing may be comparable to weight-based dosing in overall predicted prevention efficacy. INTERPRETATION These findings suggest that bnAb serum concentration may be a useful marker for dosing regimen selection, and operationally efficient fixed dosing regimens could be considered for future trials of HIV-1 bnAbs. FUNDING Was provided by the National Institutes of Health, National Institute of Allergy and Infectious Diseases (NIAID) (UM1 AI068614, to the HIV Vaccine Trials Network [HVTN]; UM1 AI068635, to the HVTN Statistical Data and Management Center [SDMC], Fred Hutchinson Cancer Center [FHCC]; 2R37 054165 to the FHCC; UM1 AI068618, to HVTN Laboratory Center, FHCC; UM1 AI068619, to the HPTN Leadership and Operations Center; UM1 AI068613, to the HIV Prevention Trials Network [HPTN] Laboratory Center; UM1 AI068617, to the HPTN SDMC; and P30 AI027757, to the Center for AIDS Research, Duke University (AI P30 AI064518) and University of Washington (P30 AI027757) Centers for AIDS Research; R37AI054165 from NIAID to the FHCC; and OPP1032144 CA-VIMC Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Kelly E Seaton
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA.
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA; Department of Global Health, University of Washington, Seattle, WA, 98195, USA.
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jack R Heptinstall
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Caroline Brackett
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Kelvin Chiong
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Lily Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Nicole L Yates
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Mark Sampson
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Erika Rudnicki
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Allan C deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James I Mullins
- Department of Global Health, University of Washington, Seattle, WA, 98195, USA; Departments of Microbiology and Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Carolyn Williamson
- Division of Medical Virology, Institute of Infectious Disease & Molecular Medicine, University of Cape Town and National Health Laboratory Service, South Africa
| | - Raabya Rossenkhan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Elena E Giorgi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Avi Kenny
- Department of Biostatistics, University of Washington, Seattle, WA, 98195, USA
| | - Heather Angier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - April Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Michelle Rojas
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Marcella Sarzotti-Kelsoe
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Lu Zhang
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Sheetal Sawant
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | | | | | | | - John Hural
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - M Julianna McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | | | | | - Jesse Clark
- Department of Medicine, Division of Infectious Disease and Department of Family Medicine in the David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Fatima Laher
- Perinatal HIV Research Unit (PHRU), Wits Health Consortium, Soweto, Johannesburg, South Africa
| | - Catherine Orrell
- Desmond Tutu Health Foundation, University of Cape Town (Institute of Infectious Disease and Molecular Medicine, and Department of Medicine), Observatory, 7925, Cape Town, South Africa
| | - Ian Frank
- Penn Center for AIDS Research, Infectious Disease Division, University of Pennsylvania, 3400 Civic Center Boulevard Building 421, Philadelphia, PA, 19104, USA
| | - Pedro Gonzales
- Asociacion Civil Impacta Salud y Educación, San Miguel Clinical Research Center, Lima, Peru
| | - Srilatha Edupuganti
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Nyaradzo Mgodi
- University of Zimbabwe-University of California San Francisco (UZ-UCSF) Collaborative Research Programme, Harare, Zimbabwe, South Africa
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA; Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA, 98195, USA; Division of Medical Virology, University of Cape Town, Anzio Road, Observatory, 7925, Cape Town, South Africa
| | - Lynn Morris
- National Institute for Communicable Diseases, National Health Laboratory Service, Johannesburg, 2192, South Africa; Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2000, South Africa; Centre for the AIDS Programme of Research in South Africa, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - David Montefiori
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA
| | - Myron S Cohen
- Institute of Global Health and Infectious Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA; Departments of Microbiology and Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Georgia D Tomaras
- Duke Center for Human Systems Immunology, Departments of Surgery, Immunology, Molecular Genetics and Microbiology, Durham, NC, 27710, USA.
| |
Collapse
|
28
|
Diamos AG, Pardhe MD, Bergeman MH, Kamzina AS, DiPalma MP, Aman S, Chaves A, Lowe K, Kilbourne J, Hogue IB, Mason HS. A self-binding immune complex vaccine elicits strong neutralizing responses against herpes simplex virus in mice. Front Immunol 2023; 14:1085911. [PMID: 37205110 PMCID: PMC10186352 DOI: 10.3389/fimmu.2023.1085911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction It has been known for over half a century that mixing an antigen with its cognate antibody in an immune complex (IC) can enhance antigen immunogenicity. However, many ICs produce inconsistent immune responses, and the use of ICs in the development new vaccines has been limited despite the otherwise widespread success of antibody-based therapeutics. To address this problem, we designed a self-binding recombinant immune complex (RIC) vaccine which mimics the larger ICs generated during natural infection. Materials and methods In this study, we created two novel vaccine candidates: 1) a traditional IC targeting herpes simplex virus 2 (HSV-2) by mixing glycoprotein D (gD) with a neutralizing antibody (gD-IC); and 2) an RIC consisting of gD fused to an immunoglobulin heavy chain and then tagged with its own binding site, allowing self-binding (gD-RIC). We characterized the complex size and immune receptor binding characteristics in vitro for each preparation. Then, the in vivo immunogenicity and virus neutralization of each vaccine were compared in mice. Results gD-RIC formed larger complexes which enhanced C1q receptor binding 25-fold compared to gD-IC. After immunization of mice, gD-RIC elicited up to 1,000-fold higher gD-specific antibody titers compared to traditional IC, reaching endpoint titers of 1:500,000 after two doses without adjuvant. The RIC construct also elicited stronger virus-specific neutralization against HSV-2, as well as stronger cross-neutralization against HSV-1, although the proportion of neutralizing antibodies to total antibodies was somewhat reduced in the RIC group. Discussion This work demonstrates that the RIC system overcomes many of the pitfalls of traditional IC, providing potent immune responses against HSV-2 gD. Based on these findings, further improvements to the RIC system are discussed. RIC have now been shown to be capable of inducing potent immune responses to a variety of viral antigens, underscoring their broad potential as a vaccine platform.
Collapse
Affiliation(s)
- Andrew G. Diamos
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | | | | | | | | | | | | | | | - Ian B. Hogue
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Hugh S. Mason
- Center for Immunotherapy, Vaccines, and Virotherapy, Biodesign Institute at Arizona State University (ASU), School of Life Sciences, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
29
|
LeVee A, Mortimer J. The Challenges of Treating Patients with Breast Cancer and Obesity. Cancers (Basel) 2023; 15:2526. [PMID: 37173991 PMCID: PMC10177120 DOI: 10.3390/cancers15092526] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Obesity is defined as a body mass index (BMI) of 30 kg/m2 or more and is associated with worse outcomes in patients with breast cancer, resulting in an increased incidence of breast cancer, recurrence, and death. The incidence of obesity is increasing, with almost half of all individuals in the United States classified as obese. Patients with obesity present with unique pharmacokinetics and physiology and are at increased risk of developing diabetes mellitus and cardiovascular disease, which leads to specific challenges when treating these patients. The aim of this review is to summarize the impact of obesity on the efficacy and toxicity of systemic therapies used for breast cancer patients, describe the molecular mechanisms through which obesity can affect systemic therapies, outline the existing American Society of Clinical Oncology (ASCO) guidelines for treating patients with cancer and obesity, and highlight additional clinical considerations for treating patients with obesity and breast cancer. We conclude that further research on the biological mechanisms underlying the obesity-breast cancer link may offer new treatment strategies, and clinicals trials that focus on the treatment and outcomes of patients with obesity and all stages of breast cancer are needed to inform future treatment guidelines.
Collapse
Affiliation(s)
- Alexis LeVee
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | | |
Collapse
|
30
|
Ter Heine R, van den Heuvel MM, Piet B, Deenen MJ, van der Wekken AJ, Hendriks LEL, Croes S, van Geel RMJM, Jansman FGA, Boshuizen RC, Franssen EJF, Smit AAJ, Dumoulin DW, Oude Munnink TH, Smit EF, Derijks HJ, van der Leest CH, Hendrikx JJMA, Moes DJAR, de Rouw N. A Systematic Evaluation of Cost-Saving Dosing Regimens for Therapeutic Antibodies and Antibody-Drug Conjugates for the Treatment of Lung Cancer. Target Oncol 2023; 18:441-450. [PMID: 37081309 DOI: 10.1007/s11523-023-00958-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND Expensive novel anticancer drugs put a serious strain on healthcare budgets, and the associated drug expenses limit access to life-saving treatments worldwide. OBJECTIVE We aimed to develop alternative dosing regimens to reduce drug expenses. METHODS We developed alternative dosing regimens for the following monoclonal antibodies used for the treatment of lung cancer: amivantamab, atezolizumab, bevacizumab, durvalumab, ipilimumab, nivolumab, pembrolizumab, and ramucirumab; and for the antibody-drug conjugate trastuzumab deruxtecan. The alternative dosing regimens were developed by means of modeling and simulation based on the population pharmacokinetic models developed by the license holders. They were based on weight bands and the administration of complete vials to limit drug wastage. The resulting dosing regimens were developed to comply with criteria used by regulatory authorities for in silico dose development. RESULTS We found that alternative dosing regimens could result in cost savings that range from 11 to 28%, and lead to equivalent pharmacokinetic exposure with no relevant increases in variability in exposure. CONCLUSIONS Dosing regimens based on weight bands and the use of complete vials to reduce drug wastage result in less expenses while maintaining equivalent exposure. The level of evidence of our proposal is the same as accepted by regulatory authorities for the approval of alternative dosing regimens of other monoclonal antibodies in oncology. The proposed alternative dosing regimens can, therefore, be directly implemented in clinical practice.
Collapse
Affiliation(s)
- Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands.
| | | | - Berber Piet
- Department of Pulmonology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital Eindhoven, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anthonie J van der Wekken
- Department of Pulmonary Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW-School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Sander Croes
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Robin M J M van Geel
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Frank G A Jansman
- Department of Clinical Pharmacy, Deventer Hospital, Deventer, The Netherlands
- Unit of PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | | | - Eric J F Franssen
- Department of Clinical Pharmacy, Onze Lieve Vrouwe Gasthuis Hospital AC, Amsterdam, The Netherlands
| | - Arthur A J Smit
- Department of Pulmonology, Onze Lieve Vrouwe Gasthuis Hospital AC, Amsterdam, The Netherlands
| | - Daphne W Dumoulin
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Thijs H Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Egbert F Smit
- Department of Pulmonology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hieronymus J Derijks
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
- Department of Pharmacy, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | | | - Jeroen J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute (NKI-AVL), Amsterdam, The Netherlands
| | - Dirk J A R Moes
- Department of Clinical Pharmacology and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nikki de Rouw
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
- Department of Clinical Pharmacy, Amphia Hospital, Breda, The Netherlands
| |
Collapse
|
31
|
Holistic View on the Structure of Immune Response: Petri Net Model. Biomedicines 2023; 11:biomedicines11020452. [PMID: 36830988 PMCID: PMC9953182 DOI: 10.3390/biomedicines11020452] [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: 12/14/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/08/2023] Open
Abstract
The simulation of immune response is a challenging task because quantitative data are scarce. Quantitative theoretical models either focus on specific cell-cell interactions or have to make assumptions about parameters. The broad variation of, e.g., the dimensions and abundance between lymph nodes as well as between individual patients hampers conclusive quantitative modeling. No theoretical model has been established representing a consensus on the set of major cellular processes involved in the immune response. In this paper, we apply the Petri net formalism to construct a semi-quantitative mathematical model of the lymph nodes. The model covers the major cellular processes of immune response and fulfills the formal requirements of Petri net models. The intention is to develop a model taking into account the viewpoints of experienced pathologists and computer scientists in the field of systems biology. In order to verify formal requirements, we discuss invariant properties and apply the asynchronous firing rule of a place/transition net. Twenty-five transition invariants cover the model, and each is assigned to a functional mode of the immune response. In simulations, the Petri net model describes the dynamic modes of the immune response, its adaption to antigens, and its loss of memory.
Collapse
|
32
|
Bittner B. Customer-centric product presentations for monoclonal antibodies. AAPS OPEN 2023; 9:3. [PMID: 36713112 PMCID: PMC9869842 DOI: 10.1186/s41120-022-00069-y] [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: 09/21/2022] [Accepted: 12/02/2022] [Indexed: 01/25/2023] Open
Abstract
Delivering customer-centric product presentations for biotherapeutics, such as monoclonal antibodies (mAbs), represents a long-standing and paramount area of engagement for pharmaceutical scientists. Activities include improving experience with the dosing procedure, reducing drug administration-related expenditures, and ultimately shifting parenteral treatments outside of a controlled healthcare institutional setting. In times of increasingly cost-constrained markets and reinforced with the coronavirus pandemic, this discipline of "Product Optimization" in healthcare has gained momentum and changed from a nice-to-have into a must. This review summarizes latest trends in the healthcare ecosystem that inform key strategies for developing customer-centric products, including the availability of a wider array of sustainable drug delivery options and treatment management plans that support dosing in a flexible care setting. Three disease area archetypes with varying degree of implementation of customer-centric concepts are introduced to highlight relevant market differences and similarities. Namely, rheumatoid arthritis and inflammatory bowel disease, multiple sclerosis, and oncology have been chosen due to differences in the availability of subcutaneously dosed and ready-to-use self-administration products for mAb medicines and their follow-on biologics. Different launch scenarios are described from a manufacturer's perspective highlighting the necessity of platform approaches. To unfold the full potential of customer-centric care, value-based healthcare provider reimbursement schemes that incentivize the efficiency of care need to be broadly implemented.
Collapse
Affiliation(s)
- Beate Bittner
- F. Hoffmann-La Roche Ltd., Global Product Strategy - Product Optimization, Grenzacher Strasse 124, CH-4070 Basel, Switzerland
| |
Collapse
|
33
|
Li L, Qu J, Song M, Zhao Q, Yang Y, Tan X, Hu Y, Li J, Lin Y, Feng H, Yao S, Keegan P, Chen M. Flat dose regimen of toripalimab based on model-informed drug development approach. Front Pharmacol 2023; 13:1069818. [PMID: 36712659 PMCID: PMC9880172 DOI: 10.3389/fphar.2022.1069818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Introduction: Flat dosing regimen has recently been approved for programmed death receptor-1 (PD-1) inhibitors including toripalimab, nivolumab and pembrolizumab. The objective of this study is to provide pharmacological evidence for a flat dosing regimen of toripalimab by assessing the efficacy and safety profile of a 240 mg Q3W flat dose relative to the currently approved 3 mg/kg Q2W. Methods: A population pharmacokinetic (PopPK) model was established based on 1,014 evaluable patients in 13 clinical studies. The exposure-objective response rate (ORR, n = 234) and exposure-safety (n = 152) analyses were performed by logistic regression. Three safety endpoints including grade ≥ 3 adverse events (AEs), treatment-related grade ≥ 3 AEs, and AEs leading to study drug discontinuation were evaluated. Progression-free survival (PFS, n = 234) was evaluated using a Cox proportional hazard model with the Kaplan-Meier survival curve. Results: The PK profiles of toripalimab are best described by a two-compartment model with time-varying clearance characterized by a sigmoidal maximum effect (Emax) function. Simulations for the first dose and steady-state exposures for the 240 mg Q3W dosing regimen were comparable to those for the 3 mg/kg Q2W dosing regimen with 95% exposure coverage ranging from 88% to 96%. The exposure-safety analysis showed that the probability of an adverse event occurring did not increase with increases in toripalimab exposure. A flat exposure-response relationship for ORR was identified. The Kaplan-Meier survival curve showed that exposure was a predictor for PFS; however, no difference in treatment benefit was demonstrated across exposure quantiles using a Cox proportional hazard model. Discussion: This study revealed that toripalimab exposure of 240 mg Q3W dosing regimen was comparable to 3 mg/kg Q2W dosing regimen. The safety and efficacy E-R results of 240 mg Q3W is flat. Hence, the 240 mg Q3W dosing regimen is determined to be a preferred therapeutic dosage for toripalimab due to the convenience of flat dose.
Collapse
Affiliation(s)
- Lili Li
- Shanghai Junshi Biosciences, Shanghai, China
| | - Jianye Qu
- Shanghai Junshi Biosciences, Shanghai, China
| | - Ming Song
- Shanghai Junshi Biosciences, Shanghai, China
| | - Qun Zhao
- Shanghai Junshi Biosciences, Shanghai, China
| | | | - Xi Tan
- Shanghai Junshi Biosciences, Shanghai, China
| | - Yanyan Hu
- Shanghai Junshi Biosciences, Shanghai, China
| | - Jing Li
- Shanghai Junshi Biosciences, Shanghai, China
| | - Yunfei Lin
- Shanghai Junshi Biosciences, Shanghai, China
| | - Hui Feng
- Shanghai Junshi Biosciences, Shanghai, China,TopAlliance Biosciences, Rockville, MD, United States
| | - Sheng Yao
- Shanghai Junshi Biosciences, Shanghai, China,TopAlliance Biosciences, Rockville, MD, United States
| | | | - Meixia Chen
- Shanghai Junshi Biosciences, Shanghai, China,*Correspondence: Meixia Chen,
| |
Collapse
|
34
|
Desai M, Kundu A, Hageman M, Lou H, Boisvert D. Monoclonal antibody and protein therapeutic formulations for subcutaneous delivery: high-concentration, low-volume vs. low-concentration, high-volume. MAbs 2023; 15:2285277. [PMID: 38013454 DOI: 10.1080/19420862.2023.2285277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
Biologic drugs are used to treat a variety of cancers and chronic diseases. While most of these treatments are administered intravenously by trained healthcare professionals, a noticeable trend has emerged favoring subcutaneous (SC) administration. SC administration of biologics poses several challenges. Biologic drugs often require higher doses for optimal efficacy, surpassing the low volume capacity of traditional SC delivery methods like autoinjectors. Consequently, high concentrations of active ingredients are needed, creating time-consuming formulation obstacles. Alternatives to traditional SC delivery systems are therefore needed to support higher-volume biologic formulations and to reduce development time and other risks associated with high-concentration biologic formulations. Here, we outline key considerations for SC biologic drug formulations and delivery and explore a paradigm shift: the flexibility afforded by low-to-moderate-concentration drugs in high-volume formulations as an alternative to the traditionally difficult approach of high-concentration, low-volume SC formulation delivery.
Collapse
Affiliation(s)
- M Desai
- Medical Affairs, Enable Injections, Inc, Cincinnati, OH, USA
| | - A Kundu
- Manufacturing Sciences, Takeda Pharmaceuticals, Brooklyn Park, MN, USA
| | - M Hageman
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, USA
| | - H Lou
- Biopharmaceutical Innovation & Optimization Center, The University of Kansas, Lawrence, KS, USA
| | - D Boisvert
- Independent Chemistry Manufacturing & Controls (CMC) Consultant, El Cerrito, CA, USA
| |
Collapse
|
35
|
Thimotheo Batista JP, Santos Marzano LA, Menezes Silva RA, de Sá Rodrigues KE, Simões E Silva AC. Chemotherapy and Anticancer Drugs Adjustment in Obesity: A Narrative Review. Curr Med Chem 2023; 30:1003-1028. [PMID: 35946096 DOI: 10.2174/0929867329666220806140204] [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: 10/18/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Obese individuals have higher rates of cancer incidence and cancer- related mortality. The worse chemotherapy outcomes observed in this subset of patients are multifactorial, including the altered physiology in obesity and its impact on pharmacokinetics, the possible increased risk of underdosing, and treatment-related toxicity. AIMS The present review aimed to discuss recent data on physiology, providing just an overall perspective and pharmacokinetic alterations in obesity concerning chemotherapy. We also reviewed the controversies of dosing adjustment strategies in adult and pediatric patients, mainly addressing the use of actual total body weight and ideal body weight. METHODS This narrative review tried to provide the best evidence to support antineoplastic drug dosing strategies in children, adolescents, and adults. RESULTS Cardiovascular, hepatic, and renal alterations of obesity can affect the distribution, metabolism, and clearance of drugs. Anticancer drugs have a narrow therapeutic range, and variations in dosing may result in either toxicity or underdosing. Obese patients are underrepresented in clinical trials that focus on determining recommendations for chemotherapy dosing and administration in clinical practice. After considering associated comorbidities, the guidelines recommend that chemotherapy should be dosed according to body surface area (BSA) calculated with actual total body weight, not an estimate or ideal weight, especially when the intention of therapy is the cure. CONCLUSION The actual total body weight dosing appears to be a better approach to dosing anticancer drugs in both adults and children when aiming for curative results, showing no difference in toxicity and no limitation in treatment outcomes compared to adjusted doses.
Collapse
Affiliation(s)
- João Pedro Thimotheo Batista
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil
| | - Lucas Alexandre Santos Marzano
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil
| | - Renata Aguiar Menezes Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil
| | - Karla Emília de Sá Rodrigues
- Departmento de Pediatria, Faculdade de Medicina, Universidade Federal de Minas Gerais, CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efgênia, Belo Horizonte, MG, Brazil
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efigênia, Belo Horizonte, MG, Brazil.,Departmento de Pediatria, Faculdade de Medicina, Universidade Federal de Minas Gerais, CEP 30.130-100, Avenida Professor Alfredo Balena, nº190/sl 281, Santa Efgênia, Belo Horizonte, MG, Brazil
| |
Collapse
|
36
|
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: 17.0] [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
|
37
|
Xu Z, Leu JH, Xu Y, Nnane I, Liva SG, Wang-Lin SX, Kudgus-Lokken R, Vermeulen A, Ouellet D. Development of Therapeutic Proteins for a New Subcutaneous Route of Administration after the Establishment of Intravenous Dosages: A Systematic Review. Clin Pharmacol Ther 2022; 113:1011-1029. [PMID: 36516352 DOI: 10.1002/cpt.2823] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Therapeutic proteins may first be developed as intravenous (i.v.) therapies with new subcutaneous (s.c.) dosage forms being subsequently developed to provide an alternative route of administration. As of August 2022, there have been 9 therapeutic proteins which were developed as a new s.c. dosage form after the approval of the corresponding i.v. product. This article provides a systematic review of prior experiences in the i.v. to s.c. switch development programs. We describe what types of clinical studies were conducted to support the i.v. to s.c. switch for these nine therapeutic proteins. Publicly available scientific advice from health authorities is summarized, particularly regarding recommendations on overall development strategy, dose selection, immunogenicity assessment, and indication extrapolation. The clinical data from these i.v. to s.c. development programs demonstrate that: (1) when switching from i.v. dosing to s.c. dosing, trough drug concentration (Ctrough ) from s.c. dosing should not be inferior to i.v. dosing with average drug concentration (Cavg ; equivalent to AUC, area under the curve after correcting for dosing intervals between i.v. and s.c. administration) being matched or non-inferior to i.v. dosing; and (2) with appropriate s.c. dose regimens, treatment with s.c. therapeutic proteins can generally achieve similar efficacy and safety as the corresponding i.v. products, suggesting that the much higher maximum concentration (Cmax ) after i.v. infusion as compared with that from s.c. injection is often not relevant to the treatment effect.
Collapse
Affiliation(s)
- Zhenhua Xu
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC., Spring House, Pennsylvania, USA
| | - Jocelyn H Leu
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC., Spring House, Pennsylvania, USA
| | - Yan Xu
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC., Spring House, Pennsylvania, USA
| | - Ivo Nnane
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC., Spring House, Pennsylvania, USA
| | - Sophia G Liva
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC., Spring House, Pennsylvania, USA
| | - Shun Xin Wang-Lin
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC., Spring House, Pennsylvania, USA
| | | | - An Vermeulen
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Daniele Ouellet
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC., Spring House, Pennsylvania, USA
| |
Collapse
|
38
|
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]
|
39
|
Combined Vaccination with B Cell Peptides Targeting Her-2/neu and Immune Checkpoints as Emerging Treatment Option in Cancer. Cancers (Basel) 2022; 14:cancers14225678. [PMID: 36428769 PMCID: PMC9688220 DOI: 10.3390/cancers14225678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/07/2022] [Accepted: 11/12/2022] [Indexed: 11/22/2022] Open
Abstract
The application of monoclonal antibodies (mAbs), targeting tumor-associated (TAAs) or tumor-specific antigens or immune checkpoints (ICs), has shown tremendous success in cancer therapy. However, the application of mAbs suffers from a series of limitations, including the necessity of frequent administration, the limited duration of clinical response and the emergence of frequently pronounced immune-related adverse events. However, the introduction of mAbs has also resulted in a multitude of novel developments for the treatment of cancers, including vaccinations against various tumor cell-associated epitopes. Here, we reviewed recent clinical trials involving combination therapies with mAbs targeting the PD-1/PD-L1 axis and Her-2/neu, which was chosen as a paradigm for a clinically highly relevant TAA. Our recent findings from murine immunizations against the PD-1 pathway and Her-2/neu with peptides representing the mimotopes/B cell peptides of therapeutic antibodies targeting these molecules are an important focus of the present review. Moreover, concerns regarding the safety of vaccination approaches targeting PD-1, in the context of the continuing immune response, as a result of induced immunological memory, are also addressed. Hence, we describe a new frontier of cancer treatment by active immunization using combined mimotopes/B cell peptides aimed at various targets relevant to cancer biology.
Collapse
|
40
|
Vaidya R, Till C, Greenlee H, Hershman DL, Unger JM. Trends in Obesity Prevalence Among Patients Enrolled in Clinical Trials for Obesity-Related Cancers, 1986 to 2016. JAMA Netw Open 2022; 5:e2234445. [PMID: 36215073 PMCID: PMC9552895 DOI: 10.1001/jamanetworkopen.2022.34445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
IMPORTANCE Obesity rates have risen in the United States since the 1980s. Several studies have shown links between obesity and the incidence of specific cancer types, but none have systematically examined obesity prevalence at the time of cancer diagnosis, or among clinical trial participants, all of which may affect clinical outcomes. OBJECTIVE To examine the prevalence of obesity and, separately, overweight and obesity over 30 years among patients with cancer enrolled in clinical treatment trials for obesity-related cancers; and to compare trends with corresponding trends in the US. DESIGN, SETTING, AND PARTICIPANTS This cohort study examined clinical treatment trials for obesity-related cancers conducted by the SWOG Cancer Research Network at community and academic sites. Participants included adult patients enrolled in phase 2 or phase 3 clinical treatment trials in obesity-related cancers between 1986 and 2016. Statistical analysis was performed from June 2020 to July 2022. EXPOSURES Year of enrollment to a clinical trial. MAIN OUTCOMES AND MEASURES Prevalence of obesity (body mass index [BMI] ≥ 30) and overweight or obesity (BMI > 25) at the time of clinical trial enrollment. Multivariable logistic regression analysis, adjusted for demographic and clinical factors, was used to analyze patient height and weight collected at clinical trial enrollment. RESULTS Among 23 926 patients (median [IQR] age, 58 [51-66] years; 17 594 [73.5%] female; 969 [4.0%] Hispanic, 2173 [9.1%] non-Hispanic Black, 19 890 [83.1%] non-Hispanic White) enrolled between 1986 and 2016, unadjusted obesity rates increased from 23.5% (in 1986 to 1990 [n = 657]) to 42.3% (in 2011 to 2016 [n = 836]). There was an increasing linear trend in obesity (odds ratio [OR], 1.23 for each 5-year increase; 95% CI, 1.21-1.26; P < .001), which persisted after covariate adjustment for demographic and clinical characteristics. Findings were consistent for the combined prevalence of overweight and obesity. The observed overall increasing trend in obesity prevalence from 1999-2000 to 2015-2016 was greater in trial patients (21.3% [SE = 0.7] to 49.1% [SE = 1.6]) than in US adults (30.5% [SE = 1.5] to 39.6% [SE = 1.6]) (P for trend = .03), but was similar to US cancer survivors (18.9% [SE = 1.9] to 42.2% [SE = 2.1]; P for trend = .31). CONCLUSIONS AND RELEVANCE This cohort study found that patients with cancer and obesity are currently well-represented in cancer clinical trials, increasing confidence that trial findings are generalizable to patients with obesity in clinical practice. Availability of data on height and weight at diagnosis through cancer registries will enable a more rigorous analysis of obesity prevalence among patients with cancer.
Collapse
Affiliation(s)
- Riha Vaidya
- SWOG Statistics and Data Management Center, Seattle, Washington
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Cathee Till
- SWOG Statistics and Data Management Center, Seattle, Washington
- Fred Hutchinson Cancer Center, Seattle, Washington
| | | | | | - Joseph M. Unger
- SWOG Statistics and Data Management Center, Seattle, Washington
- Fred Hutchinson Cancer Center, Seattle, Washington
| |
Collapse
|
41
|
Olivier T, Haslam A, Prasad V. Dose modification rules and availability of growth factor support: A cross-sectional study of head-to-head cancer trials used for US FDA approval from 2009 to 2021. Eur J Cancer 2022; 172:349-356. [PMID: 35830842 DOI: 10.1016/j.ejca.2022.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022]
Abstract
AIM OF THE STUDY Different drug modification rules or growth factor support guidance may affect the results in oncology randomised controlled trials. We aimed to estimate the prevalence of unequal rules for dose modification rules or the use of myeloid growth factors in head-to-head registration Food and Drug Administration trials. METHODS This cross-sectional analysis included all head-to-head registration randomised controlled trials leading to a US Food and Drug Administration approval between 2009 and 2021. Trials examined anti-cancer drugs in the advanced or metastatic setting where a comparison could be made between arms regarding either dose modification rules or myeloid growth factors recommendations. Sixty-two registration trials met inclusion criteria. Information abstracted for each trial included tumour type, setting, phase, and type of sponsor. We assessed, according to pre-specified rules, imbalance in drug modification rules, myeloid growth factors recommendations or both. RESULTS We find 40 of 62 (65%) selected trials have unequal rules for dose medication, granulocyte colony-stimulating factor (G-CSF) use or both. Six trials (10%) had rules favouring the control arm, while 55% of selected trials (34/62) favoured the experimental arm. Among these, 50% (17/34) had unequal drug modification rules, 41% (14/34) had unequal G-CSF rules and 9% contained both (3/34). CONCLUSION We find that 55% of trials testing anti-cancer drugs against each other used protocol rules that favoured the experimental arm. This leaves open the question of whether new molecules are truly superior to older molecules or if instead different outcomes are due to more aggressive dosing or growth factor support. Trials should utilise equal rules for dose medication and G-CSF support.
Collapse
Affiliation(s)
- Timothée Olivier
- Department of Oncology, Geneva University Hospital, 4 Gabrielle-Perret-Gentil Street, 1205, Geneva, Switzerland; Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA.
| | - Alyson Haslam
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA
| |
Collapse
|
42
|
Tobias J, Drinić M, Högler S, Ambroz K, Baier K, Kodajova P, Tomasich E, Berghoff AS, Schmid A, Garner-Spitzer E, Kenner L, Kundi M, Zielinski CC, Wiedermann U. Active immunization with a Her-2/neu-targeting Multi-peptide B cell vaccine prevents lung metastases formation from Her-2/neu breast cancer in a mouse model. Transl Oncol 2022; 19:101378. [PMID: 35259675 PMCID: PMC8904231 DOI: 10.1016/j.tranon.2022.101378] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022] Open
Abstract
Trastuzumab and Pertuzumab extend the overall survival of breast cancer patients. Unlike monoclonal antibodies B cell peptides induce immunological memory. A multi-peptide B cell-based vaccination prevents Her-2/neu lung metastasis in mice. The vaccination results in Her-2/neu-negative tumors, with increased PD-L1 expression. A combination/alternating therapy for a total remission of metastases is suggested.
In pre-clinical and clinical settings, active immunization with a Her-2/neu vaccine (HerVaxx), comprising B-cell peptide from Trastuzumab binding site, has been shown to reduce primary tumor growth via induction of polyclonal anti-tumor immune responses and immunological memory. Here, we tested the combination of HerVaxx and the recently identified B-cell epitope/mimotope of Pertuzumab, i.e. a multi-peptide B-cell vaccine, for preventing Her-2/neu lung metastases formation in a mouse model. Active immunization with the multi-peptide vaccine was associated with decreased lung weights, and histological evaluation of the lungs showed that the significant reduction of lung metastases was associated with increased CD4+ and CD8+ T cell infiltration. Notably, along with the overall reduction of lungs weights and Her-2 positive metastases, a formation of Her-2/neu-negative tumors but with increased PD-L1 expression was observed. Our results might pave the way to a multi-peptide B-cell Her-2/neu vaccine serving as a secondary intervention in adjuvant settings to prevent tumor recurrence and spread. Moreover, combination therapy targeting PD-L1 may result in total remission of metastases. Such a therapy may be used clinically to alternately target Her-2/neu and PD-L1 in metastatic breast cancer.
Collapse
|
43
|
Abstract
Biosimilars have come of age over the past 17 years, with 84 approvals in the EU and 35 in the US, representing almost 90% of the world market. While the acceptance of biosimilars in the US is catching up with that in the EU, the cost benefits remain elusive due to the high development barrier and complex distribution system involved, mainly in the US. In the EU, the cost of biosimilars has already dropped 70% or more, and interchangeability is a routine in some European jurisdictions, unlike in the US, where a separate regulatory approval is required. This paper projects significant changes coming in the US and EU’s biosimilars approval requirements that will impact the approval procedures in the rest of the world, leading to dramatic changes in the cost of biosimilars to patients. This perspective is based on the author’s first-hand experience to secure FDA approvals of biosimilars and an extensive analysis of the rationality of testing to demonstrate biosimilarity. Multiple citizen petitions by the author and meetings with the FDA may have prompted the recent announcement by the FDA to award a $5 million research grant to scientists to develop novel testing models to establish biosimilarity, including modifying the interchangeability protocols. Soon, demonstration of biosimilarity will not require animal testing and, in most cases, clinical efficacy testing; over time, the clinical pharmacology testing will be reduced as the regulatory agencies develop more confidence in the safety and efficacy of biosimilars. Biosimilars have come of age; now it is the turn of the developers to grow up, and one way to show this is to challenge the current regulatory guidelines but only on scientific grounds to seek more concessions, for which both FDA and EMA are ready.
Collapse
|
44
|
Masters JC, Khandelwal A, di Pietro A, Dai H, Brar S. Model-informed drug development supporting the approval of the avelumab flat-dose regimen in patients with advanced renal cell carcinoma. CPT Pharmacometrics Syst Pharmacol 2022; 11:458-468. [PMID: 35166465 PMCID: PMC9007597 DOI: 10.1002/psp4.12771] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 01/24/2022] [Indexed: 11/29/2022] Open
Abstract
Avelumab is an anti–PD‐L1 monoclonal antibody approved as monotherapy for Merkel cell carcinoma (MCC) and urothelial carcinoma (UC), and in combination with axitinib for advanced renal cell carcinoma (aRCC). Although initially approved with weight‐based dosing (10 mg/kg intravenously [IV] every 2 weeks [Q2W]), avelumab was subsequently approved for flat dosing (800 mg IV Q2W) based on population pharmacokinetic (PopPK), exposure‐efficacy, and exposure‐safety modeling in MCC and UC. Here, through modeling and simulation, we provide justification for a flat‐dose regimen of avelumab plus axitinib in aRCC. Simulated exposure metrics from the previous monotherapy PopPK model (1827 patients) for both weight‐based and flat‐dose regimens were compared with exposure metrics from treatment‐naive patients with aRCC who received avelumab plus axitinib (488 patients). The aRCC population exposures were derived from a fit‐for‐purpose PopPK model developed using data from monotherapy and combination studies and the existing base structural PopPK model. Exposure‐response relationships for safety were analyzed, including grade ≥3 treatment‐emergent adverse events (TEAEs), any‐grade infusion‐related reactions, and TEAE any‐grade immune‐related adverse events (irAEs). Weight‐based dosing of avelumab in the aRCC population yielded similar PK exposures to the flat‐dose regimen reference exposures in the monotherapy population. Increased avelumab exposure was not associated with increased probabilities of grade ≥3 TEAEs or any‐grade IRRs, although there was a weak association with an increased probability of any‐grade irAEs. Overall, models in aRCC suggest that the avelumab 800‐mg Q2W flat‐dose regimen would provide similar benefits compared with weight‐based dosing with no meaningful change in the probability of AEs.
Collapse
|
45
|
Papandreou P, Nousiou K, Papandreou G, Steier J, Skouroliakou M, Karageorgopoulou S. The use of a novel clinical decision support system for reducing medication errors and expediting care in the provision of chemotherapy. HEALTH AND TECHNOLOGY 2022. [DOI: 10.1007/s12553-022-00654-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
46
|
de Jong KA, Rosing H, Huitema AD, Beijnen JH. Optimized sample pre-treatment procedure for the simultaneous UPLC-MS/MS quantification of ipilimumab, nivolumab, and pembrolizumab in human serum. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1196:123215. [DOI: 10.1016/j.jchromb.2022.123215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 11/30/2022]
|
47
|
Gamain B, Brousse C, Rainey NE, Diallo BK, Paquereau CE, Desrames A, Ceputyte J, Semblat JP, Bertrand O, Gangnard S, Teillaud JL, Chêne A. BMFPs, a versatile therapeutic tool for redirecting a preexisting Epstein-Barr virus antibody response toward defined target cells. SCIENCE ADVANCES 2022; 8:eabl4363. [PMID: 35148183 PMCID: PMC8836820 DOI: 10.1126/sciadv.abl4363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
Industrial production of therapeutic monoclonal antibodies is mostly performed in eukaryotic-based systems, allowing posttranslational modifications mandatory for their functional activity. The resulting elevated product cost limits therapy access to some patients. To address this limitation, we conceptualized a novel immunotherapeutic approach to redirect a preexisting polyclonal antibody response against Epstein-Barr virus (EBV) toward defined target cells. We engineered and expressed in bacteria bimodular fusion proteins (BMFPs) comprising an Fc-deficient binding moiety targeting an antigen expressed at the surface of a target cell, fused to the EBV-P18 antigen, which recruits circulating endogenous anti-P18 IgG in EBV+ individuals. Opsonization of BMFP-coated targets efficiently triggered antibody-mediated clearing effector mechanisms. When assessed in a P18-primed mouse tumor model, therapy performed with an anti-huCD20 BMFP significantly led to increased survival and total cancer remission in some animals. These results indicate that BMFPs could represent potent and useful therapeutic molecules to treat a number of diseases.
Collapse
Affiliation(s)
- Benoît Gamain
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Carine Brousse
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Nathan E. Rainey
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Béré K. Diallo
- Laboratory “Immune Microenvironment and Immunotherapy”, INSERM U.1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Faculté de Médecine, Sorbonne Université, 91 boulevard de l’Hôpital, 75013 Paris, France
| | - Clara-Eva Paquereau
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Alexandra Desrames
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Jolita Ceputyte
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Jean-Philippe Semblat
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Olivier Bertrand
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Stéphane Gangnard
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| | - Jean-Luc Teillaud
- Laboratory “Immune Microenvironment and Immunotherapy”, INSERM U.1135, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Faculté de Médecine, Sorbonne Université, 91 boulevard de l’Hôpital, 75013 Paris, France
| | - Arnaud Chêne
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, INSERM, F-75015 Paris, France
| |
Collapse
|
48
|
Al‑Saleh K, Abdel‑Warith A, Alghamdi M, Aldiab A, Ali A, Alsaeed E, Abozeed W, Abdel‑aziz N. Incidence of trastuzumab‑induced cardiotoxicity and impact of body mass index in patients with breast cancer: Results from a Saudi tertiary cancer center. Mol Clin Oncol 2022; 16:78. [DOI: 10.3892/mco.2022.2511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 05/05/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Khalid Al‑Saleh
- Department of Medicine, College of Medicine, King Saud University, Riyadh 12372, Saudi Arabia
| | - Ahmed Abdel‑Warith
- Division of Hematology/Oncology, Department of Medicine, King Khalid University Hospital, College of Medicine, King Saud University Medical City, Riyadh 12372, Saudi Arabia
| | - Mohammed Alghamdi
- Division of Hematology/Oncology, Department of Medicine, King Khalid University Hospital, College of Medicine, King Saud University Medical City, Riyadh 12372, Saudi Arabia
| | - Abdurrahman Aldiab
- Division of Hematology/Oncology, Department of Medicine, King Khalid University Hospital, College of Medicine, King Saud University Medical City, Riyadh 12372, Saudi Arabia
| | - Arwa Ali
- Department of Medical Oncology, South Egypt Cancer Institute, Assiut University, Assiut 71516, Egypt
| | - Eyad Alsaeed
- Department of Oncology, Division of Radiation Oncology, King Khalid University Hospital, College of Medicine, King Saud University Medical City, Riyadh 12372, Saudi Arabia
| | - Waleed Abozeed
- Clinical Oncology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Nashwa Abdel‑aziz
- Division of Hematology/Oncology, Department of Medicine, King Khalid University Hospital, College of Medicine, King Saud University Medical City, Riyadh 12372, Saudi Arabia
| |
Collapse
|
49
|
Maritaz C, Broutin S, Chaput N, Marabelle A, Paci A. Immune checkpoint-targeted antibodies: a room for dose and schedule optimization? J Hematol Oncol 2022; 15:6. [PMID: 35033167 PMCID: PMC8760805 DOI: 10.1186/s13045-021-01182-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-CTLA-4 and anti-PD-1/PD-L1 immune checkpoint inhibitors are therapeutic monoclonal antibodies that do not target cancer cells but are designed to reactivate or promote antitumor immunity. Dosing and scheduling of these biologics were established according to conventional drug development models, even though the determination of a maximum tolerated dose in the clinic could only be defined for anti-CTLA-4. Given the pharmacology of these monoclonal antibodies, their high interpatient pharmacokinetic variability, the actual clinical benefit as monotherapy that is observed only in a specific subset of patients, and the substantial cost of these treatments, a number of questions arise regarding the selected dose and the dosing interval. This review aims to outline the development of these immunotherapies and considers optimization options that could be used in clinical practice.
Collapse
Affiliation(s)
- Christophe Maritaz
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sophie Broutin
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Chaput
- Laboratory for Immunomonitoring in Oncology (LIO), Faculty of Pharmacy, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Aurélien Marabelle
- Drug Development Unit (DITEP), LRTI U1015 INSERM, Gustave Roussy, Villejuif, France
| | - Angelo Paci
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France.
- Pharmacokinetic Unit, Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.
| |
Collapse
|
50
|
Tobias J, Garner-Spitzer E, Drinić M, Wiedermann U. Vaccination against Her-2/neu, with focus on peptide-based vaccines. ESMO Open 2022; 7:100361. [PMID: 35026721 PMCID: PMC8760406 DOI: 10.1016/j.esmoop.2021.100361] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy has been a milestone in combatting cancer, by complementing or even replacing classic treatments like surgery, chemotherapy, radiation, and anti-hormonal therapy. In 15%-30% of breast cancers, overexpression of the human epidermal growth factor receptor 2 (Her-2/neu) is associated with more aggressive tumor development. Passive immunization/immunotherapy with the recombinantly produced Her-2/neu-targeting monoclonal antibodies (mAbs) pertuzumab and trastuzumab has been shown to effectively treat breast cancer and lead to a significantly better prognosis. However, allergic and hypersensitivity reactions, cardiotoxicity, development of resistance, lack of immunological memory which results in continuous application over a long period, and cost-intensiveness are among the drawbacks associated with this treatment. Furthermore, intrinsic or acquired resistance is associated with the application of therapeutic mAbs, leading to the disease recurrence. Conversely, these drawbacks could be potentially overcome by vaccination, i.e. an active immunization/immunotherapy approach by activating the patient’s own immune system to target cancer, along with inducing immunological memory. This review aims to summarize the main approaches investigated and undertaken for the production of Her-2/neu vaccine candidates, with the main focus on peptide-based vaccines and their evaluation in clinical settings. Her-2/neu is overexpressed in 10%-30% of breast and gastric cancer patients and this correlates with poor clinical outcomes. Passive application of trastuzumab and pertuzumab has outstandingly improved the Her-2/neu-related clinical outcomes. Treatment with mAbs is associated with frequent administration, cost-intensiveness, and resistance. Vaccination against Her-2/neu with e.g. mimotope- or peptide-based vaccines can alternatively overcome the mAbs’ drawbacks. Such alternatives may pave the way to therapeutics which could be used as monotherapy or in combination therapies with mAbs.
Collapse
Affiliation(s)
- J Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| | - E Garner-Spitzer
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - M Drinić
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - U Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|