1
|
Sicard G, Protzenko D, Giacometti S, Barlési F, Ciccolini J, Fanciullino R. Harnessing tumor immunity with cytotoxics: T cells monitoring in mice bearing lung tumors treated with anti-VEGF and pemetrexed-cisplatin doublet. Br J Cancer 2023; 129:1373-1382. [PMID: 37524968 PMCID: PMC10628115 DOI: 10.1038/s41416-023-02350-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/19/2023] [Accepted: 06/27/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Successful immunotherapy is restricted to some cancers only, and combinatorial strategies with other drugs could help to improve their efficacy. Here, we monitor T cells in NSCLC model after treatment with cytotoxics (CT) and anti-VEGF drugs, to understand when immune checkpoint inhibitors should be best associated next. METHODS In vivo study was performed on BALB/c mice grafted with KLN205 cells. Eight treatments were tested including control, cisplatin and pemetrexed as low (LD CT) and full (MTD CT) dose as single agents, flat dose anti-VEGF and the association anti-VEGF + CT. Full immunomonitoring was performed by flow cytometry on tumor, spleen and blood over 3 weeks. RESULTS Immunomodulatory effect was dependent upon both treatments and time. In tumors, combination groups shown numerical lower Treg cells on Day 21. In spleen, anti-VEGF and LD CT group shown higher CD8/Treg ratio on Day 7; on Day 14, higher T CD4 were observed in both combination groups. Finally, in blood, Tregs were lower and CD8/Treg ratio higher, on Day 14 in both combination groups. On Day 21, CD4 and CD8 T cells were higher in the anti-VEGF + MTD CT group. CONCLUSIONS Anti-VEGF associated to CT triggers notable increase in CD8/Tregs ratio. Regarding the scheduling, a two-week delay after using anti-VEGF and CT could be the best sequence to optimize antitumor efficacy.
Collapse
Affiliation(s)
- G Sicard
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - D Protzenko
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - S Giacometti
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - F Barlési
- School of Medicine, Aix Marseille University, 13007, Marseille, France
- Gustave Roussy Institute, 94800, Villejuif, France
| | - J Ciccolini
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France.
| | - R Fanciullino
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| |
Collapse
|
2
|
Sato M, Maishi N, Hida Y, Yanagawa-Matsuda A, Alam MT, Sakakibara-Konishi J, Nam JM, Onodera Y, Konno S, Hida K. Angiogenic inhibitor pre-administration improves the therapeutic effects of immunotherapy. Cancer Med 2023; 12:9760-9773. [PMID: 36808261 PMCID: PMC10166916 DOI: 10.1002/cam4.5696] [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: 08/10/2022] [Revised: 12/01/2022] [Accepted: 02/03/2023] [Indexed: 02/22/2023] Open
Abstract
In lung cancer, immune checkpoint inhibitors (ICIs) are often inadequate for tumor growth inhibition. Angiogenic inhibitors (AIs) are required to normalize tumor vasculature for improved immune cell infiltration. However, in clinical practice, ICIs and cytotoxic antineoplastic agents are simultaneously administered with an AI when tumor vessels are abnormal. Therefore, we examined the effects of pre-administering an AI for lung cancer immunotherapy in a mouse lung cancer model. Using DC101, an anti-vascular endothelial growth factor receptor 2 (VEGFR2) monoclonal antibody, a murine subcutaneous Lewis lung cancer (LLC) model was used to determine the timing of vascular normalization. Microvessel density (MVD), pericyte coverage, tissue hypoxia, and CD8-positive cell infiltration were analyzed. The effects of an ICI and paclitaxel after DC101 pre-administration were investigated. On Day 3, increased pericyte coverage and alleviated tumor hypoxia represented the highest vascular normalization. CD8+ T-cell infiltration was also highest on Day 3. When combined with an ICI, DC101 pre-administration significantly reduced PD-L1 expression. When combined with an ICI and paclitaxel, only DC101 pre-administration significantly inhibited tumor growth, but simultaneous administration did not. AI pre-administration, and not simultaneous administration, may increase the therapeutic effects of ICIs due to improved immune cell infiltration.
Collapse
Affiliation(s)
- Mineyoshi Sato
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan.,Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Nako Maishi
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan.,Advanced Robotic and Endoscopic Surgery, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Aya Yanagawa-Matsuda
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Mohammad Towfik Alam
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| | - Jun Sakakibara-Konishi
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Jin-Min Nam
- Global Center for Biomedical Science and Engineering (GCB), Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering (GCB), Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Kyoko Hida
- Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
3
|
Muñoz R, Girotti A, Hileeto D, Arias FJ. Metronomic Anti-Cancer Therapy: A Multimodal Therapy Governed by the Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13215414. [PMID: 34771577 PMCID: PMC8582362 DOI: 10.3390/cancers13215414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Metronomic chemotherapy with different mechanisms of action against cancer cells and their microenvironment represents an exceptional holistic cancer treatment. Each type of tumor has its own characteristics, including each individual tumor in each patient. Understanding the complexity of the dynamic interactions that take place between tumor and stromal cells and the microenvironment in tumor progression and metastases, as well as the response of the host and the tumor itself to anticancer therapy, will allow therapeutic actions with long-lasting effects to be implemented using metronomic regimens. This study aims to highlight the complexity of cellular interactions in the tumor microenvironment and summarize some of the preclinical and clinical results that explain the multimodality of metronomic therapy, which, together with its low toxicity, supports an inhibitory effect on the primary tumor and metastases. We also highlight the possible use of nano-therapeutic agents as good partners for metronomic chemotherapy. Abstract The concept of cancer as a systemic disease, and the therapeutic implications of this, has gained special relevance. This concept encompasses the interactions between tumor and stromal cells and their microenvironment in the complex setting of primary tumors and metastases. These factors determine cellular co-evolution in time and space, contribute to tumor progression, and could counteract therapeutic effects. Additionally, cancer therapies can induce cellular and molecular responses in the tumor and host that allow them to escape therapy and promote tumor progression. In this study, we describe the vascular network, tumor-infiltrated immune cells, and cancer-associated fibroblasts as sources of heterogeneity and plasticity in the tumor microenvironment, and their influence on cancer progression. We also discuss tumor and host responses to the chemotherapy regimen, at the maximum tolerated dose, mainly targeting cancer cells, and a multimodal metronomic chemotherapy approach targeting both cancer cells and their microenvironment. In a combination therapy context, metronomic chemotherapy exhibits antimetastatic efficacy with low toxicity but is not exempt from resistance mechanisms. As such, a better understanding of the interactions between the components of the tumor microenvironment could improve the selection of drug combinations and schedules, as well as the use of nano-therapeutic agents against certain malignancies.
Collapse
Affiliation(s)
- Raquel Muñoz
- Department of Biochemistry, Physiology and Molecular Biology, University of Valladolid, Paseo de Belén, 47011 Valladolid, Spain
- Smart Biodevices for NanoMed Group, University of Valladolid, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
- Correspondence:
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
| | - Denise Hileeto
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON N2L 361, Canada;
| | - Francisco Javier Arias
- Smart Biodevices for NanoMed Group, University of Valladolid, LUCIA Building, Paseo de Belén, 47011 Valladolid, Spain;
| |
Collapse
|
4
|
Avallone A, Piccirillo MC, Nasti G, Rosati G, Carlomagno C, Di Gennaro E, Romano C, Tatangelo F, Granata V, Cassata A, Silvestro L, De Stefano A, Aloj L, Vicario V, Nappi A, Leone A, Bilancia D, Arenare L, Petrillo A, Lastoria S, Gallo C, Botti G, Delrio P, Izzo F, Perrone F, Budillon A. Effect of Bevacizumab in Combination With Standard Oxaliplatin-Based Regimens in Patients With Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2118475. [PMID: 34309665 PMCID: PMC8314140 DOI: 10.1001/jamanetworkopen.2021.18475] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IMPORTANCE Although bevacizumab is a standard of care in combination treatments for metastatic colorectal cancer (mCRC), its clinical benefit has been limited. OBJECTIVE To determine whether sequential scheduling of bevacizumab administration in combination with chemotherapy improves treatment efficacy in patients with mCRC, in keeping with the tumor vascular normalization hypothesis. DESIGN, SETTING, AND PARTICIPANTS This open-label, randomized clinical phase 3 trial was conducted from May 8, 2012, to December 9, 2015, at 3 Italian centers. Patients aged 18 to 75 years with unresectable, previously untreated, or single line-treated mCRC were recruited. Follow-up was completed December 31, 2019, and data were analyzed from February 26 to July 24, 2020. INTERVENTIONS Patients received 12 biweekly cycles of standard oxaliplatin-based regimens (modified FOLFOX-6 [levo-folinic acid, fluorouracil, and oxaliplatin]/modified CAPOX [capecitabine and oxaliplatin]) plus bevacizumab administered either on the same day as chemotherapy (standard arm) or 4 days before chemotherapy (experimental arm). MAIN OUTCOMES AND MEASURES The primary end point was the objective response rate (ORR) measured with Response Evaluation Criteria in Solid Tumors, version 1.1. Secondary end points included progression-free survival, overall survival, safety, and quality of life (QOL). RESULTS Overall, 230 patients (136 men [59.1%]; median age, 62.3 [interquartile range, 53.3-67.6] years) were randomly assigned to the standard arm (n = 115) or the experimental arm (n = 115). The median duration of follow-up was 68.3 (95% CI, 61.0-70.0) months. No difference in ORR (57.4% [95% CI, 47.8%-66.6%] in the standard arm and 56.5% [95% CI, 47.0-65.7] in the experimental arm; P = .89) or progression-free survival (10.5 [95% CI, 9.1-12.3] months in the standard arm and 11.7 [95% CI, 9.9-12.9] months in the experimental arm; P = .15) was observed. However, the median overall survival was 29.8 (95% CI, 22.5-41.1) months in the experimental arm compared with 24.1 (95% CI, 18.6-29.8) months in the standard arm (adjusted hazard ratio, 0.73; 95% CI, 0.54-0.99; P = .04). Moreover, the experimental arm was associated with a significant reduction in the rate of severe diarrhea (6 [5.3%] vs 19 [16.5%]; P = .006) and nausea (2 [1.8%] vs 8 [7.0%]; P = .05) and improved physical functioning (mean [SD] change from baseline, 0.65 [1.96] vs -7.41 [2.95] at 24 weeks; P = .02), and constipation scores (mean [SD] change from baseline, -17.2 [3.73] vs -0.62 [4.44]; P = .003). CONCLUSIONS AND RELEVANCE In this randomized clinical trial, sequential administration of bevacizumab plus chemotherapy did not improve ORR, the primary end point. However, the overall survival advantage, fewer adverse effects, and better health-related QOL associated with sequential bevacizumab administration might provide the basis for exploring antiangiogenic combination treatments with innovative perspectives. TRIAL REGISTRATION EudraCT Identifier: 2011-004997-27; ClinicalTrials.gov Identifier: NCT01718873.
Collapse
Affiliation(s)
- Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Maria C. Piccirillo
- Clinical Trials Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Guglielmo Nasti
- Innovative Therapy for Abdominal Metastases, IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Gerardo Rosati
- Medical Oncology Unit, S. Carlo Hospital, Potenza, Italy
| | - Chiara Carlomagno
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Carmela Romano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Fabiana Tatangelo
- Pathology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Vincenza Granata
- Radiology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Antonino Cassata
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Lucrezia Silvestro
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Alfonso De Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Luigi Aloj
- Nuclear Medicine Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
- currently affiliated with Department of Radiology, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Valeria Vicario
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Anna Nappi
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione G. Pascale, Napoli, Italy
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | | | - Laura Arenare
- Clinical Trials Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Antonella Petrillo
- Radiology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Secondo Lastoria
- Nuclear Medicine Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Ciro Gallo
- Università della Campania Luigi Vanvitelli, Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Paolo Delrio
- Colorectal Oncological Surgery, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Francesco Izzo
- Colorectal Oncological Surgery, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
- Hepatobiliary Surgery Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Franco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori–IRCCS, Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
5
|
Poels KE, Schoenfeld AJ, Makhnin A, Tobi Y, Wang Y, Frisco-Cabanos H, Chakrabarti S, Shi M, Napoli C, McDonald TO, Tan W, Hata A, Weinrich SL, Yu HA, Michor F. Identification of optimal dosing schedules of dacomitinib and osimertinib for a phase I/II trial in advanced EGFR-mutant non-small cell lung cancer. Nat Commun 2021; 12:3697. [PMID: 34140482 PMCID: PMC8211846 DOI: 10.1038/s41467-021-23912-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 05/24/2021] [Indexed: 02/03/2023] Open
Abstract
Despite the clinical success of the third-generation EGFR inhibitor osimertinib as a first-line treatment of EGFR-mutant non-small cell lung cancer (NSCLC), resistance arises due to the acquisition of EGFR second-site mutations and other mechanisms, which necessitates alternative therapies. Dacomitinib, a pan-HER inhibitor, is approved for first-line treatment and results in different acquired EGFR mutations than osimertinib that mediate on-target resistance. A combination of osimertinib and dacomitinib could therefore induce more durable responses by preventing the emergence of resistance. Here we present an integrated computational modeling and experimental approach to identify an optimal dosing schedule for osimertinib and dacomitinib combination therapy. We developed a predictive model that encompasses tumor heterogeneity and inter-subject pharmacokinetic variability to predict tumor evolution under different dosing schedules, parameterized using in vitro dose-response data. This model was validated using cell line data and used to identify an optimal combination dosing schedule. Our schedule was subsequently confirmed tolerable in an ongoing dose-escalation phase I clinical trial (NCT03810807), with some dose modifications, demonstrating that our rational modeling approach can be used to identify appropriate dosing for combination therapy in the clinical setting.
Collapse
Affiliation(s)
- Kamrine E Poels
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
| | - Adam J Schoenfeld
- Division of Solid Tumor Oncology, Department of Medicine, Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Alex Makhnin
- Division of Solid Tumor Oncology, Department of Medicine, Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Yosef Tobi
- Division of Solid Tumor Oncology, Department of Medicine, Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Yuli Wang
- Oncology Research and Development, Pfizer Inc, La Jolla, CA, USA
| | | | - Shaon Chakrabarti
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Manli Shi
- Oncology Research and Development, Pfizer Inc, La Jolla, CA, USA
| | - Chelsi Napoli
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Thomas O McDonald
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- The Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Weiwei Tan
- Clinical Pharmacology Oncology, Global Product Development, Pfizer Inc, San Diego, CA, USA
| | - Aaron Hata
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
- The Ludwig Center at Harvard, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Scott L Weinrich
- Oncology Research and Development, Pfizer Inc, La Jolla, CA, USA
| | - Helena A Yu
- Division of Solid Tumor Oncology, Department of Medicine, Thoracic Oncology Service, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| | - Franziska Michor
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- The Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA, USA.
- The Ludwig Center at Harvard, Boston, MA, USA.
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
6
|
Physiologically-Based Pharmacokinetic/Pharmacodynamic Model of MBQ-167 to Predict Tumor Growth Inhibition in Mice. Pharmaceutics 2020; 12:pharmaceutics12100975. [PMID: 33076517 PMCID: PMC7602742 DOI: 10.3390/pharmaceutics12100975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/02/2020] [Accepted: 10/11/2020] [Indexed: 01/01/2023] Open
Abstract
MBQ-167 is a dual inhibitor of the Rho GTPases Rac and Cdc42 that has shown promising results as an anti-cancer therapeutic at the preclinical stage. This drug has been tested in vitro and in vivo in metastatic breast cancer mouse models. The aim of this study is to develop a physiologically based pharmacokinetic/pharmacodynamic (PBPK-PD) model of MBQ-167 to predict tumor growth inhibition following intraperitoneal (IP) administration in mice bearing Triple Negative and HER2+ mammary tumors. PBPK and Simeoni tumor growth inhibition (TGI) models were developed using the Simcyp V19 Animal Simulator. Our developed PBPK framework adequately describes the time course of MBQ-167 in each of the mouse tissues (e.g., lungs, heart, liver, kidneys, spleen, plasma) and tumor, since the predicted results were consistent with the experimental data. The developed PBPK-PD model successfully predicts tumor shrinkage in HER2+ and triple-negative breast tumors after the intraperitoneal administration of 1 and 10 mg/kg body weight (BW) dose level of MBQ-167 three times a week. The findings from this study suggest that MBQ-167 has a higher net effect and potency inhibiting Triple Negative mammary tumor growth compared to HER2+ and that liver metabolism is the major route of elimination of this drug.
Collapse
|
7
|
Ferrer F, Fanciullino R, Milano G, Ciccolini J. Towards Rational Cancer Therapeutics: Optimizing Dosing, Delivery, Scheduling, and Combinations. Clin Pharmacol Ther 2020; 108:458-470. [PMID: 32557660 DOI: 10.1002/cpt.1954] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/30/2020] [Indexed: 12/16/2022]
Abstract
The current trend to personalize anticancer therapies mostly relies on selecting the best drug or combination of drugs to achieve optimal efficacy in patients. In addition to the comprehensive genetic and molecular knowledge of each tumor before choosing the drugs to be given, there is probably much room left for improvement by further personalizing the very modes by which the drugs are given, once they have been carefully selected. In particular, shifting from standard dosing to tailored dosing should help in maintaining drug exposure levels in the right therapeutic window, thus ensuring that the efficacy/toxicity balance is optimal. This paper covers the current knowledge regarding pharmacokinetic/pharmacodynamic relationships of anticancer agents, from decades-old cytotoxics to the latest immune checkpoint inhibitors, the most frequent sources for long-neglected interpatient variability impacting on drug exposure levels, and what could be done to achieve real personalized medicine in oncology such as implementing therapeutic drug monitoring with adaptive dosing strategies or using model-driven modalities for personalized dosing and scheduling.
Collapse
Affiliation(s)
- Florent Ferrer
- SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France
| | | | - Gérard Milano
- Onco-Pharmacology Unit, Centre Antoine Lacassagne, Nice, France
| | - Joseph Ciccolini
- SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France
| |
Collapse
|