1
|
Shivanna AT, Dash BS, Lu YJ, Lin WT, Chen JP. Magnetic lipid-poly(lactic-co-glycolic acid) nanoparticles conjugated with epidermal growth factor receptor antibody for dual-targeted delivery of CPT-11. Int J Pharm 2024; 667:124856. [PMID: 39461680 DOI: 10.1016/j.ijpharm.2024.124856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/01/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
To entrap sparingly water-soluble drugs like CPT-11 (irinotecan), the poly(lactic-co-glycolic acid) (PLGA) nanoparticle (NP) is highly favored due to its low cytotoxicity and approval for clinical use. On the other hand, entrapping hydrophobic oleic acid-coated iron oxide magnetic nanoparticles (OMNP) in PLGA NP can provide a nanovehicle for magnetically targeted drug delivery. Our goal in this study is to develop a new dual-targeted magnetic lipid-polymer NP for the delivery of CPT-11. We first co-entrap OMNP and CPT-11 in self-assembled lipid-PLGA NP to prepare OLNP@CPT-11. The OLNP@CPT-11 surface was modified with an epidermal growth factor receptor (EGFR) antibody Cetuximab (CET), which can actively target the overexpressed EGFR on the U87 glioblastoma cell surface. The OLNP-CET@CPT-11 enables dual targeting through both external magnetic guidance and CET-mediated active targeting. The NP was characterized for physicochemical properties using various analytical techniques. In vitro study confirms ligand-receptor interaction results in enhanced endocytosis of OLNP-CET@CPT-11 by U87 cells, which offers increased cytotoxicity and elevated cell apoptosis rates. Furthermore, magnetic guidance of OLNP-CET@CPT-11 to U87 cells can induce cell death exclusively in the magnetically targeted zone. The dual-targeted strategy also provides the best therapeutic efficacy against subcutaneously implanted U87 tumors in nude mice with intravenously delivered OLNP-CET@CPT-11.
Collapse
Affiliation(s)
- Anilkumar T Shivanna
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan
| | - Wei-Ting Lin
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Kwei-San, Taoyuan 33305, Taiwan; Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 33302, Taiwan; Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
2
|
Nandi S, Swain R, Habibullah S, Sahoo RN, Nayak AK, Mallick S. Lipid-Gelucire based rectal delivery of ramipril prodrug exhibits significant lowering of intra-ocular pressure in normotensive rabbit: sustained structural relaxation release kinetics and IVIVC. Pharm Dev Technol 2024; 29:468-476. [PMID: 38662798 DOI: 10.1080/10837450.2024.2345807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
Carboxylesterase enzymes convert a prodrug ramipril into the biologically active metabolite ramiprilat. It is prescribed for controlling ocular hypertension after oral administration. High concentrations of carboxylesterase enzymes in rectal and colon tissue can transform ramipril significantly to ramiprilat. Sustained rectal delivery of ramipril has been developed for intra-ocular pressure lowering effect using a normotensive rabbit model. Rectal suppositories have been formulated using a matrix base of HPMC K100-PEG 400-PEG 6000, incorporating varying amounts of Gelucire by the fusion moulding method. The presence of Gelucire in the suppository exhibited sustained structural relaxation-based release kinetics of RM compared to its absence. Intravenous and oral administration of ramipril has decreased IOP in the treated rabbit up to 90 and 360 min, respectively. Treated rabbits with suppositories have revealed decreased IOP for an extended period compared to the above. Formulation containing GEL 3% reduced intra-ocular pressure to 540 min, with the highest area under the decreased IOP curve. Compared to oral, the pharmacodynamic bioavailability of ramipril has been improved significantly using a sustained-release rectal suppository. A rectal suppository for sustained delivery of ramipril could be used to lower IOP significantly.
Collapse
Affiliation(s)
- Souvik Nandi
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Rakesh Swain
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Sk Habibullah
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Rudra Narayan Sahoo
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Amit Kumar Nayak
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Subrata Mallick
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| |
Collapse
|
3
|
de With M, van Doorn L, Kloet E, van Veggel A, Matic M, de Neijs MJ, Oomen-de Hoop E, van Meerten E, van Schaik RHN, Mathijssen RHJ, Bins S. Irinotecan-Induced Toxicity: A Pharmacogenetic Study Beyond UGT1A1. Clin Pharmacokinet 2023; 62:1589-1597. [PMID: 37715926 PMCID: PMC10582127 DOI: 10.1007/s40262-023-01279-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND AND OBJECTIVE Side effects of irinotecan treatment can be dose limiting and may impair quality of life. In this study, we investigated the correlation between single nucleotide polymorphisms (SNPs) in genes encoding enzymes involved in the irinotecan metabolism and transport, outside UGT1A1, and irinotecan-related toxicity. We focused on carboxylesterases, which are involved in formation of the active metabolite SN-38 and on drug transporters. METHODS Patients who provided written informed consent at the Erasmus Medical Center Cancer Institute to the Code Geno study (local protocol: MEC02-1002) or the IRI28-study (NTR-6612) were enrolled in the study and were genotyped for 15 SNPs in the genes CES1, CES2, SLCO1B1, ABCB1, ABCC2, and ABCG2. RESULTS From 299 evaluable patients, 86 patients (28.8%) developed severe irinotecan-related toxicity. A significantly higher risk of toxicity was seen in ABCG2 c.421C>A variant allele carriers (P = 0.030, OR 1.88, 95% CI 1.06-3.34). Higher age was associated with all grade diarrhea (P = 0.041, OR 1.03, 95% CI 1.00-1.06). In addition, CES1 c.1165-41C>T and CES1 n.95346T>C variant allele carriers had a lower risk of all-grade thrombocytopenia (P = 0.024, OR 0.42, 95% CI 0.20-0.90 and P = 0.018, OR 0.23, 95% CI 0.08-0.79, respectively). CONCLUSION Our study indicates that ABCG2 and CES1 SNPs might be used as predictive markers for irinotecan-induced toxicity.
Collapse
Affiliation(s)
- Mirjam de With
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Clinical Chemistry, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Leni van Doorn
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Esmay Kloet
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Anne van Veggel
- Department of Clinical Chemistry, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Maja Matic
- Department of Clinical Chemistry, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Micha J de Neijs
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Wang Z, Li W, Park J, Gonzalez KM, Scott AJ, Lu J. Camptothesome elicits immunogenic cell death to boost colorectal cancer immune checkpoint blockade. J Control Release 2022; 349:929-939. [PMID: 35926754 DOI: 10.1016/j.jconrel.2022.07.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/22/2022]
Abstract
Camptothesome is an innovative nanovesicle therapeutic comprising the sphingomyelin-derived camptothecin (CPT) lipid bilayer. In this work, we deciphered that Camptothesome was taken up by colorectal cancer (CRC) cells through primarily the clathrin-mediated endocytotic pathway and displayed the potential of eliciting robust immunogenic cancer cell death (ICD) via upregulating calreticulin, high mobility group box 1 protein (HMGB-1), and adenosine triphosphate (ATP), three hallmarks involved in the induction of ICD. In addition, use of dying MC38 tumor cells treated with Camptothesome as vaccine prevented tumor growth in 60% mice that received subsequent injection of live MC38 cells on the contralateral flank, validating Camptothesome was a legitimate ICD inducer in vivo. Camptothesome markedly reduced the acute bone marrow toxicity and gastrointestinal mucositis associated with free CPT and beat free CPT and Onivyde on anti-CRC efficacy and immune responses in a partially interferon gamma (IFN-γ)-dependent manner. Furthermore, Camptothesome enhanced the efficacy of immune checkpoint inhibitors to shrink late-stage orthotopic MC38 CRC tumors with diminished tumor metastasis and markedly prolonged mice survival.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Jonghan Park
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States
| | - Aaron James Scott
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ 85721, United States; Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, United States; Southwest Environmental Health Sciences Center, The University of Arizona, Tucson 85721, United States.
| |
Collapse
|
5
|
Milano G, Innocenti F, Minami H. Liposomal irinotecan (Onivyde): Exemplifying the benefits of nanotherapeutic drugs. Cancer Sci 2022; 113:2224-2231. [PMID: 35445479 PMCID: PMC9277406 DOI: 10.1111/cas.15377] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022] Open
Abstract
Irinotecan is a topoisomerase inhibitor, widely used in treatment of malignancies including pancreatic ductal adenocarcinoma (PDAC) as part of the FOLFIRINOX regimen prescribed as a first-line treatment in several countries. However, irinotecan has not been successfully introduced as a second-line treatment for pancreatic cancer and few randomized clinical studies have evaluated its added value. Efficacy of liposomal irinotecan (nal-IRI) combined with 5-fluorouracil and leucovorin (5-FU/LV) was reported in the phase III NAPOLI-1 trial in metastatic PDAC following failure of gemcitabine-based therapy. Several features of nal-IRI pharmacokinetics (PK) could result in better outcomes versus nonliposomal irinotecan. Irinotecan is a prodrug that is converted to active SN-38 by carboxylesterase enzymes and inactivated by cytochrome P450 3A4/3A5. SN-38 is inactivated by UGT1A1 enzymes. Individual variations in their expression and activity could influence enhanced localized irinotecan activity and toxicity. Liposomal irinotecan exploits the enhanced permeability and retention effect in cancer, accumulating in tumor tissues. Liposomal irinotecan also has a longer half-life and higher area under the concentration-time curve (0-∞) than nonliposomal irinotecan, as the liposomal formulation protects cargo from premature metabolism in the plasma. This results in irinotecan activation in tumor tissue, leading to enhanced cytotoxicity. Importantly, despite the longer exposure, overall toxicity for nal-IRI is no worse than nonliposomal irinotecan. Liposomal irinotecan exemplifies how liposomal encapsulation of a chemotherapeutic agent can alter its PK properties, improving clinical outcomes for patients. Liposomal irinotecan is currently under investigation in other malignancies including biliary tract cancer (amongst other gastrointestinal cancers), brain tumors, and small-cell lung cancer.
Collapse
Affiliation(s)
- Gérard Milano
- UPR 7497Scientific Valorisation UnitCentre Antoine Lacassagne and Côte d’Azur UniversityNiceFrance
| | | | - Hironobu Minami
- Medical Oncology and HematologyKobe University Graduate School of Medicine and HospitalKobeJapan
| |
Collapse
|
6
|
Kailass K, Sadovski O, Zipfel WR, Beharry AA. Two-Photon Photodynamic Therapy Targeting Cancers with Low Carboxylesterase 2 Activity Guided by Ratiometric Fluorescence. J Med Chem 2022; 65:8855-8868. [PMID: 35700557 DOI: 10.1021/acs.jmedchem.1c01965] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human carboxylesterase 2 (hCES2) converts anticancer prodrugs, such as irinotecan, into their active metabolites via phase I drug metabolism. Owing to interindividual variability, hCES2 serves as a predictive marker of patient response to hCES2-activated prodrug-based therapy, whereby a low intratumoral hCES2 activity leads to therapeutic resistance. Despite the ability to identify nonresponders, effective treatments for resistant patients are needed. Clinically approved photodynamic therapy is an attractive alternative for irinotecan-resistant patients. Here, we describe the application of our hCES2-selective small-molecule ratiometric fluorescent chemosensor, Benz-AP, as a single theranostic agent given its discovered functionality as a photosensitizer. Benz-AP produces singlet oxygen and induces photocytotoxicity in cancer cells in a strong negative correlation with hCES2 activity. Two-photon excitation of Benz-AP produces fluorescence, singlet oxygen, and photocytotoxicity in tumor spheroids. Overall, Benz-AP serves as a novel theranostic agent with selective photocytotoxicity in hCES2-prodrug resistant cancer cells, making Benz-AP a promising agent for in vivo applications.
Collapse
Affiliation(s)
- Karishma Kailass
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Oleg Sadovski
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Warren R Zipfel
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Andrew A Beharry
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| |
Collapse
|
7
|
Wagner S, Beger NT, Matschos S, Szymanski A, Przybylla R, Bürtin F, Prall F, Linnebacher M, Mullins CS. Tumour-Derived Cell Lines and Their Potential for Therapy Prediction in Patients with Metastatic Colorectal Cancer. Cancers (Basel) 2021; 13:4717. [PMID: 34572946 PMCID: PMC8471446 DOI: 10.3390/cancers13184717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/10/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
The prognosis of metastatic colorectal cancer (CRC) remains poor. Patients and physicians are in need of individual therapies and precise response predictions. We investigated the predictive capacity of primary tumour material for treatment response of metastases. Mutational landscapes of primary tumours and corresponding metastases of 10 CRC patients were compared. Cell line characteristics and chemosensitivity were investigated pairwise for primary and metastatic tumours of four patients. PDX models of one patient were treated in vivo for proof of concept. Driver mutations did not differ between primaries and metastases, while the latter accumulated additional mutations. In vitro chemosensitivity testing revealed no differences for responses to 5-FU and oxaliplatin between primary and metastatic cell lines. However, irinotecan response differed significantly: the majority of metastases-derived cell lines was less sensitive to irinotecan than their matching primary counterpart. Therapy recommendations based on these findings were compared to clinical treatment response and mostly in line with the predicted outcome. Therefore, primary tumour cell models seem to be a good tool for drug response testing and conclusion drawing for later metastases. With further data from tumour-derived cell models, such predictions could improve clinical treatment decisions, both recommending likely effective therapeutic options while excluding ineffective treatments.
Collapse
Affiliation(s)
- Sandra Wagner
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| | - Nicola T. Beger
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| | - Stephanie Matschos
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| | - Antonia Szymanski
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| | - Randy Przybylla
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| | - Florian Bürtin
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| | - Friedrich Prall
- Institute of Pathology, University Medicine Rostock, 18057 Rostock, Germany;
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| | - Christina S. Mullins
- Molecular Oncology and Immunotherapy, Department of General Surgery, University Medicine Rostock, 18057 Rostock, Germany; (S.W.); (N.T.B.); (S.M.); (A.S.); (R.P.); (F.B.); (C.S.M.)
| |
Collapse
|
8
|
Melancon MP, Yevich S, Avritscher R, Swigost A, Lu L, Tian L, Damasco JA, Dixon K, Cortes AC, Munoz NM, Liang D, Liu D, Tam AL. A novel irinotecan-lipiodol nanoemulsion for intravascular administration: pharmacokinetics and biodistribution in the normal and tumor bearing rat liver. Drug Deliv 2021; 28:240-251. [PMID: 33501859 PMCID: PMC8725905 DOI: 10.1080/10717544.2020.1869863] [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] [Indexed: 11/17/2022] Open
Abstract
Colorectal cancer is one of the most common cancers in the United States and treatment options are limited for patients who develop liver metastases. Several chemotherapeutic regimens have been used for transvascular liver-directed therapy in the treatment of colorectal liver metastases without clear evidence of superiority of one therapy over another. We describe the development of a novel nanoemulsion through combining irinotecan (IRI), a first line systemic agent used for the treatment of colon cancer, with lipiodol, an oily contrast medium derived from poppy seed oil, and evaluated its pharmacokinetic and biodistribution profile as a function of portal venous chemoembolization (PVCE) versus transarterial chemoembolization (TACE) delivery. The Tessari technique was used to create a stable emulsion (20 mg IRI mixed with 2 mL lipiodol) with resultant particle size ranging from 28.9 nm to 56.4 nm. Pharmacokinetic profile established through venous sampling in Buffalo rats demonstrate that the area under the curve (AUC0−∞) of IRI was significantly less after PVCE with IRI-lipiodol as compared to IRI alone (131 vs. 316 µg*min/mL, p-value = .023), suggesting significantly higher amounts of IRI retention in the liver with the IRI-lipiodol nanoemulsion via first-pass extraction. Subseqent biodistribution studies in tumor-bearing WAG/Rjj rats revealed more IRI present in the tumor following TACE versus PVCE (29.19 ± 12.33 µg/g versus 3.42 ± 1.62; p-value = .0033) or IV (29.19 ± 12.33 µg/g versus 1.05 ± 0.47; p-value = .0035). The IRI-lipiodol nanoemulsion demonstrated an acceptable hepatotoxicity profile in all routes of administration. In conclusion, the IRI-lipiodol nanoemulsion via TACE showed promise and warrants further investigation as an option for the treatment of metastatic colorectal cancer.
Collapse
Affiliation(s)
- Marites P Melancon
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas MD Anderson Cancer Center UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Steven Yevich
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rony Avritscher
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adam Swigost
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Medstar Georgetown University Hospital, Washington Hospital Center, Washington, DC, USA
| | - Linfeng Lu
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Baylor College of Medicine, Houston, TX, USA
| | - Li Tian
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jossana A Damasco
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katherine Dixon
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrea C Cortes
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nina M Munoz
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dong Liang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA
| | - David Liu
- The University of British Columbia, Vancouver, Canada
| | - Alda L Tam
- Department of Interventional Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
9
|
Heterogenous chemosensitivity of a panel of organoid lines derived from small cell neuroendocrine carcinoma of the uterine cervix. Hum Cell 2021; 34:889-900. [PMID: 33677798 DOI: 10.1007/s13577-021-00511-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/15/2021] [Indexed: 10/22/2022]
Abstract
Small cell neuroendocrine carcinoma (SCNEC) of the uterine cervix is a rare disease with a poor prognosis. The lack of established disease models has hampered therapy development. We generated a panel of cancer tissue-originated spheroid (CTOS) lines derived from SCNEC of the uterine cervix using a method based upon cell-cell contact throughout the preparation and culturing processes. Using 11 CTOS lines, we assessed the sensitivity of various drugs used in clinical practice. Drug sensitivity assays revealed significant heterogeneous inter-CTOS chemosensitivity. Microarray analyses were then performed to identify sensitivity-related gene signatures. Specific gene sets were identified which likely contribute to the sensitivity to the tested drugs. We identified a line (Cerv54) that was exceptionally sensitive to irinotecan. Cerv54 had increased levels of CES1, which catalyzes the conversion of irinotecan to the active form, SN38, although in Cerv54 cells, SN38 was undetectable, CES1 expression and activity were markedly low compared to the liver, and a CES1 inhibitor had no effect on irinotecan sensitivity. These results suggested a novel irinotecan mode of action in Cerv54. Our CTOS lines may be useful for understanding the variation and mechanism of drug sensitivity, contributing to the understanding and development of chemotherapeutic drugs.
Collapse
|
10
|
Pengwei G, Song Q, Li T, Cao L, Tang H, Wang Y, Tu P, Zheng J, Song Y, Li J. Confirmative Structural Annotation for Metabolites of ( R)-7,3'-Dihydroxy-4'-methoxy-8-methylflavane, A Natural Sweet Taste Modulator, by Liquid Chromatography-Three-Dimensional Mass Spectrometry. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12454-12466. [PMID: 33084329 DOI: 10.1021/acs.jafc.0c05154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Flavonoids occupy the largest family of natural products and possess a broad spectrum of health benefits. Their metabolites are sometimes the truly effective molecules in vivo. It is still challenging, however, to unambiguously identify flavonoid metabolites using conventional LC-MS/MS. Herein, we aimed to pursue auxiliary structural clues to m/z values in both MS1 and MS2 spectra through LC coupled to three-dimensional MS (LC-3D MS). MS1, as the first dimension, was in charge of suggesting theoretical molecular formulas, MS2, the as second dimension, was responsible for offering substructures, and exactly, online energy-resolved MS (ER-MS), as the third dimension, provided optimal collision energies (OCEs) that reflected the linkage manners among the substructures. Metabolic characterization of a natural sweet taste modulator, namely, (R)-7,3'-dihydroxy-4'-methoxy-8-methylflavane (DHMMF), was conducted as a proof-of-concept. Extensive efforts, such as full MS1 and MS2 scans on IT-TOF-MS and predictive selected-reaction monitoring mode on Qtrap-MS, were made for in-depth metabolite mining. Seventeen metabolites (M1-M17) were captured from DHMMF-treated biological samples, including 17 (M1-M17), 10 (M4-M9, M11, M13, M14, and M16), and 2 (M5 and M10) metabolites from urine, plasma, and feces, respectively. Their structures were configured by integrating MS1, MS2, and OCE information. Except M10, all metabolites were new compounds. LC-MS/MS-guided chromatographic purification yielded three glucuronyl-conjugated metabolites (M5, M8, and M11), and NMR spectroscopic assays consolidated the structures transmitted from LC-3D MS. Demethylation, glucuronidation, and sulfation occurred as the primary metabolic pathways of DHMMF. Above all, LC-3D MS bridged LC-MS/MS from putatively structural annotation toward confidence-enhanced identification, beyond the metabolite characterization of flavonoids.
Collapse
Affiliation(s)
- Guan Pengwei
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qingqing Song
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ting Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Libo Cao
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Huiting Tang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiao Zheng
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuelin Song
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
11
|
Mikhail AS, Levy EB, Krishnasamy VP, Woods DL, Esparza-Trujillo JA, Bakhutashvili I, Banovac F, Wakim PG, Negussie AH, Tang Y, Henman A, Willis SL, Karanian JW, Pritchard WF, Lewis AL, Wood BJ. Safety and Tolerability of Topotecan-Eluting Radiopaque Microspheres for Hepatic Chemoembolization in a Rabbit Preclinical Model. Cardiovasc Intervent Radiol 2020; 43:1918-1924. [PMID: 32803282 DOI: 10.1007/s00270-020-02609-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE Topotecan is a camptothecin analogue with potential advantages over irinotecan for transarterial chemoembolization (TACE) of hepatic colorectal metastases including greater anti-neoplastic activity without enzymatic activation. The purpose of this study was to assess safety and tolerability of topotecan-loaded radiopaque microspheres (ROMTOP) administered by TACE in a rabbit model and to compare the in vitro elution of topotecan from microspheres to irinotecan. MATERIALS AND METHODS Topotecan was loaded into radiopaque microspheres (70-150 µm, DC Bead LUMI™, Biocompatibles UK Ltd-Boston Scientific Corporation) to the maximum capacity of 80 mg/mL of microspheres. Six healthy New Zealand White rabbits underwent hepatic TACE with ROMTOP under fluoroscopic guidance until angiographic stasis. Assessment of toxicities included regular liver function tests and complete blood counts until euthanasia 28 days post-TACE. In vitro topotecan elution from the microspheres was assessed using an open-loop flow-through system and compared to irinotecan. RESULTS The mean bead volume and topotecan dose delivered were 0.086 mL (0.076-0.105 mL) and 1.99 mg/kg (1.51-2.55 mg/kg), respectively. Aspartate aminotransferase and alanine aminotransferase were elevated post-embolization but resolved within 2 weeks. One rabbit died two days after TACE with pyloric duodenal perforation observed at necropsy, potentially due to non-target embolization. In vitro elution of topotecan from ROMTOP was complete in 10 h compared to 3 h for irinotecan-loaded microspheres. CONCLUSION Selective embolization with ROMTOP was tolerated at a dose of 2 mg/kg (24 mg/m2) in rabbits. In vitro topotecan elution from microspheres was more prolonged compared to irinotecan.
Collapse
Affiliation(s)
- Andrew S Mikhail
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA.
| | - Elliot B Levy
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Venkatesh P Krishnasamy
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - David L Woods
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Juan A Esparza-Trujillo
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Ivane Bakhutashvili
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Filip Banovac
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Paul G Wakim
- Biostatistics and Clinical Epidemiology Service, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Ayele H Negussie
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Yiqing Tang
- Biocompatibles UK Ltd (a BTG International Group Company), Lakeview, Riverside Way, Watchmoor Park, Camberley, GU15 3YL, Surrey, UK
| | - Alexander Henman
- Biocompatibles UK Ltd (a BTG International Group Company), Lakeview, Riverside Way, Watchmoor Park, Camberley, GU15 3YL, Surrey, UK
| | - Sean L Willis
- Biocompatibles UK Ltd (a BTG International Group Company), Lakeview, Riverside Way, Watchmoor Park, Camberley, GU15 3YL, Surrey, UK
| | - John W Karanian
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - William F Pritchard
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| | - Andrew L Lewis
- Biocompatibles UK Ltd (a BTG International Group Company), Lakeview, Riverside Way, Watchmoor Park, Camberley, GU15 3YL, Surrey, UK
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center, National Institutes of Health, 10 Center Dr, Bethesda, MD, 20892, USA
| |
Collapse
|
12
|
Macaire P, Paris J, Vincent J, Ghiringhelli F, Bengrine-Lefevre L, Schmitt A. Impact of granulocyte colony-stimulating factor on FOLFIRINOX-induced neutropenia prevention: A population pharmacokinetic/pharmacodynamic approach. Br J Clin Pharmacol 2020; 86:2473-2485. [PMID: 32386071 DOI: 10.1111/bcp.14356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/21/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS Granulocyte colony-stimulating factor (G-CSF) is frequently prescribed to prevent chemotherapy-induced neutropenia, but the administration schedule remains empirical in case of bimonthly chemotherapy such as FOLFIRINOX regimen. This pharmacokinetic/pharmacodynamic (PK/PD) study was performed to determine the effect of different G-CSF regimens on the incidence and duration of neutropenia following FOLFIRINOX administration in order to propose an optimal G-CSF dosing schedule. METHODS A population PK/PD model was developed to describe individual neutrophil time course from absolute neutrophil counts (ANC) obtained in 40 advanced cancer patients receiving FOLFIRINOX regimen. The structural model considered ANC dynamics, neutropenic effect of cytotoxics and the stimulating effect of G-CSF on neutrophils. Final model estimates were used to simulate different G-CSF dosing schedules for 1000 virtual subjects. The incidence and duration of neutropenia were then calculated for different G-CSF dosing schedules. RESULTS The final model successfully described the myelosuppressive effect induced by the 3 cytotoxics for all patients. Simulations showed that pegfilgrastim administration reduced the risk of severe neutropenia by 22.9% for subjects with low ANC at the start of chemotherapy. Median duration in this group was also shortened by 3.1 days when compared to absence of G-CSF. Delayed G-CSF administration was responsible for higher incidence and longer duration of neutropenia compared to absence of administration. CONCLUSION The PK/PD model well described our population's ANC data. Simulations showed that pegylated-G-CSF administration 24 hours after the end of chemotherapy seems to be the optimal schedule to reduce FOLFIRINOX-induced neutropenia. We also underline the potential negative effect of G-CSF maladministration.
Collapse
Affiliation(s)
- Pauline Macaire
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| | - Justine Paris
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| | - Julie Vincent
- Oncology Department, Centre Georges-François Leclerc, Dijon, France
| | - François Ghiringhelli
- INSERM U1231, University of Burgundy Franche-Comté, Dijon, France.,Oncology Department, Centre Georges-François Leclerc, Dijon, France
| | | | - Antonin Schmitt
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| |
Collapse
|
13
|
de Man FM, Goey AKL, van Schaik RHN, Mathijssen RHJ, Bins S. Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics. Clin Pharmacokinet 2019. [PMID: 29520731 PMCID: PMC6132501 DOI: 10.1007/s40262-018-0644-7] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Since its clinical introduction in 1998, the topoisomerase I inhibitor irinotecan has been widely used in the treatment of solid tumors, including colorectal, pancreatic, and lung cancer. Irinotecan therapy is characterized by several dose-limiting toxicities and large interindividual pharmacokinetic variability. Irinotecan has a highly complex metabolism, including hydrolyzation by carboxylesterases to its active metabolite SN-38, which is 100- to 1000-fold more active compared with irinotecan itself. Several phase I and II enzymes, including cytochrome P450 (CYP) 3A4 and uridine diphosphate glucuronosyltransferase (UGT) 1A, are involved in the formation of inactive metabolites, making its metabolism prone to environmental and genetic influences. Genetic variants in the DNA of these enzymes and transporters could predict a part of the drug-related toxicity and efficacy of treatment, which has been shown in retrospective and prospective trials and meta-analyses. Patient characteristics, lifestyle and comedication also influence irinotecan pharmacokinetics. Other factors, including dietary restriction, are currently being studied. Meanwhile, a more tailored approach to prevent excessive toxicity and optimize efficacy is warranted. This review provides an updated overview on today’s literature on irinotecan pharmacokinetics, pharmacodynamics, and pharmacogenetics.
Collapse
Affiliation(s)
- Femke M de Man
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands
| | - Andrew K L Goey
- Department of Hospital Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Chamseddine AN, Ducreux M, Armand JP, Paoletti X, Satar T, Paci A, Mir O. Intestinal bacterial β-glucuronidase as a possible predictive biomarker of irinotecan-induced diarrhea severity. Pharmacol Ther 2019; 199:1-15. [DOI: 10.1016/j.pharmthera.2019.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
Sawdon AJ, Zhang J, Wang X, Peng CA. Enhanced Anticancer Activity of 5'-DFUR-PCL-MPEG Polymeric Prodrug Micelles Encapsulating Chemotherapeutic Drugs. NANOMATERIALS 2018; 8:nano8121041. [PMID: 30551585 PMCID: PMC6315712 DOI: 10.3390/nano8121041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
The compound 5’-deoxy-5-fluorouridine (5’-DFUR) is a prodrug of the anti-tumor drug 5-fluorouracil (5-FU). Thymidine phosphorylase (TP) is an enzyme that can convert 5’-DFUR to its active form 5-FU and the expression of TP is upregulated in various cancer cells. In this study, 5’-DFUR associated with amphiphilic copolymer poly(ε-caprolactone)-methoxy poly(ethylene glycol) (5’-DFUR-PCL-MPEG) was synthesized, characterized, and self-assembled into functional polymeric micelles. To demonstrate that the prodrug 5’-DFUR could convert into cytotoxic 5-fluorouracil (5-FU) by endogenous TP, HT-29 colorectal cancer cells were treated with 5’-DFUR-PCL-MPEG polymeric micelles for various time periods. Chemotherapeutic drugs doxorubicin (DOX) and 7-ethyl-10-hydroxycamptothecin (SN-38) were also encapsulated separately into 5’-DFUR-PCL-MPEG polymeric micelles to create a dual drug-loaded system. HT-29 cells were treated with DOX or SN-38 encapsulated 5’-DFUR-PCL-MPEG polymeric micelles to examine the efficacy of dual drug-loaded micelles. As a result, HT-29 cells treated with 5’-DFUR-PCL-MPEG polymeric micelles showed up to 40% cell death rate after a 72-h treatment. In contrast, HT-29 cells challenged with DOX or SN-38 encapsulated 5’-DFUR-incorporated polymeric micelles showed 36% and 31% in cell viability after a 72-h treatment, respectively.
Collapse
Affiliation(s)
- Alicia J Sawdon
- Department of Chemical Engineering, Michigan Technological University, Houghton, MI, 49931, USA.
| | - Jun Zhang
- Department of Biological Engineering, University of Idaho, Moscow, ID 83844, USA.
| | - Xutu Wang
- Department of Biological Engineering, University of Idaho, Moscow, ID 83844, USA.
| | - Ching-An Peng
- Department of Biological Engineering, University of Idaho, Moscow, ID 83844, USA.
| |
Collapse
|
16
|
Hamaguchi T, Tsuji A, Yamaguchi K, Takeda K, Uetake H, Esaki T, Amagai K, Sakai D, Baba H, Kimura M, Matsumura Y, Tsukamoto T. A phase II study of NK012, a polymeric micelle formulation of SN-38, in unresectable, metastatic or recurrent colorectal cancer patients. Cancer Chemother Pharmacol 2018; 82:1021-1029. [PMID: 30284603 PMCID: PMC6267673 DOI: 10.1007/s00280-018-3693-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022]
Abstract
Purpose NK012 is a polymeric micelle formulation of SN-38, the active metabolite of irinotecan. We evaluated the efficacy and safety of NK012 in Japanese patients with unresectable metastatic colorectal cancer. Methods We conducted a multicenter open-label phase II trial of NK012 monotherapy in 58 patients who had been treated with an oxaliplatin-based chemotherapy regimen (group A: 53 patients with UGT1A1 genotype –/–, *6/–, or *28/–; group B: 5 patients with UGT1A1 genotype *6/*28 or *6/*6). The primary endpoint was the response rate (RR). Initial doses of 28 and 18 mg/m2 for group A and group B, respectively, were administered intravenously over 30 min, and these doses were subsequently administered every 3 weeks. Group A was evaluated as the primary efficacy population, while group B was evaluated for reference. Results In group A, the RR was 3.8%, and the median progression-free survival and overall survival were 3.30 months and 15.03 months, respectively. In both groups, the most common grade ≥ 3 adverse drug reaction (ADR) was neutropenia and the incidence of grade ≥ 3 diarrhea was low or zero. In group A, 17 serious ADRs were observed in 10 patients (17%); all improved or recovered. In group B, no serious ADRs were observed. No treatment-related deaths were reported in either group. Conclusions NK012 monotherapy yielded an RR similar to the RR of irinotecan monotherapy that was reported in the phase III EPIC trial (4.2%), and the incidence of grade ≥ 3 diarrhea was low. Based on the incidence and severity of febrile neutropenia and grade ≥ 3 neutropenia, the initial dose of NK012 28 mg/m2 may be too high for colorectal cancer patients who have previously been treated with an oxaliplatin-based chemotherapy regimen.
Collapse
Affiliation(s)
- Tetsuya Hamaguchi
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan.
- Department of Gastroenterological Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, Japan.
| | - Akihito Tsuji
- Division of Medical Oncology, Kochi Health Science Center, Kochi, Japan
- Department of Clinical Oncology, Kagawa University Faculty of Medicine Cancer Center, Kagawa University Hospital, Kagawa, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
- Department of Gastroenterological Chemotherapy Center, Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Koji Takeda
- Departmentof Medical Oncology, Osaka City General Hospital, Osaka, Japan
- Approved Specified Nonprofit Corporation West Japan Oncology Group, Osaka, Japan
| | - Hiroyuki Uetake
- Department of Specialized Surgeries, Tokyo Medical and Dental University, Tokyo, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Kyushu Cancer Center, Fukuoka, Japan
| | - Kenji Amagai
- Department of Gastroenterology, Ibaraki Prefectural Central Hospital, Ibaraki, Japan
| | - Daisuke Sakai
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka, Japan
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masami Kimura
- Department of Surgery, Japan Community Health Care Organization, Hitoyoshi Medical Center, Kumamoto, Japan
| | - Yasuhiro Matsumura
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | | |
Collapse
|
17
|
Pires AS, Marques CR, Encarnação JC, Abrantes AM, Marques IA, Laranjo M, Oliveira R, Casalta-Lopes JE, Gonçalves AC, Sarmento-Ribeiro AB, Botelho MF. Ascorbic Acid Chemosensitizes Colorectal Cancer Cells and Synergistically Inhibits Tumor Growth. Front Physiol 2018; 9:911. [PMID: 30083105 PMCID: PMC6064950 DOI: 10.3389/fphys.2018.00911] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is continuously classified as one of the most incidental and mortal types of cancer worldwide. The positive outcomes of the conventional chemotherapy are frequently associated with high toxicity, which often leads to the suspension of the treatment. Growing evidences consider the use of pharmacological concentrations of ascorbic acid (AA), better known as vitamin C, in the treatment of cancer. The use of AA in a clinical context is essentially related to the adoption of new therapeutic strategies based on combination regimens, where AA plays a chemosensitizing role. The reduced sensitivity of some tumors to chemotherapy and the highly associated adverse effects continue to be some of the major obstacles in the effective treatment of CRC. So, this paper aimed to study the potential of a new therapeutic approach against this neoplasia with diminished side effects for the patient. This approach was based on the study of the combination of high concentrations of AA with reduced concentrations of drugs conventionally used in CRC patients and eligible for first and second line chemotherapeutic regimens, namely 5-fluorouracilo (5-FU), oxaliplatin (Oxa) or irinotecan (Iri). The evaluation of the potential synergy between the compounds was first assessed in vitro in three CRC cell lines with different genetic background and later in vivo using one xenograft animal model of CRC. AA and 5-FU act synergistically in vitro just for longer incubation times, however, in vivo showed no benefit compared to 5-FU alone. In contrast to the lack of synergy seen in in vitro studies with the combination of AA with irinotecan, the animal model revealed the therapeutic potential of this combination. AA also potentiated the effect of Oxa, since a synergistic effect was demonstrated, in almost all conditions and in the three cell lines. Moreover, this combined therapy (CT) caused a stagnation of the tumor growth rate, being the most promising tested combination. Pharmacological concentrations of AA increased the efficacy of Iri and Oxa against CRC, with promising results in cell lines with more aggressive phenotypes, namely, tumors with mutant or null P53 expression and tumors resistant to chemotherapy.
Collapse
Affiliation(s)
- Ana S Pires
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cláudia R Marques
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - João C Encarnação
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana M Abrantes
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Inês A Marques
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Mafalda Laranjo
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Rui Oliveira
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Department of Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - João E Casalta-Lopes
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana C Gonçalves
- Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Oncobiology and Hematology Laboratory, Applied Molecular Biology and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana B Sarmento-Ribeiro
- Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Oncobiology and Hematology Laboratory, Applied Molecular Biology and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Department of Hematology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Maria F Botelho
- Biophysics Institute, CNC.IBILI, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Institute for Clinical and Biomedical Research Area of Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
18
|
Evans JP, Winiarski BK, Sutton PA, Jones RP, Ressel L, Duckworth CA, Pritchard DM, Lin ZX, Fretwell VL, Tweedle EM, Costello E, Goldring CE, Copple IM, Park BK, Palmer DH, Kitteringham NR. The Nrf2 inhibitor brusatol is a potent antitumour agent in an orthotopic mouse model of colorectal cancer. Oncotarget 2018; 9:27104-27116. [PMID: 29930754 PMCID: PMC6007465 DOI: 10.18632/oncotarget.25497] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/07/2018] [Indexed: 12/30/2022] Open
Abstract
Nrf2 is a transcription factor that regulates cellular stress response and irinotecan-metabolising pathways. Its aberrant activity has been reported in a number of cancers, although relatively few studies have explored a role for Nrf2 in colorectal cancer (CRC). This study assessed the expression of Nrf2 in patient CRC tissues and explored the effect of Nrf2 modulation alone, or in combination with irinotecan, in human (HCT116) and murine (CT26) cell lines in vitro and in an orthotopic syngeneic mouse model utilising bioluminescent imaging. Using a tissue microarray, Nrf2 was found to be overexpressed (p<0.01) in primary CRC and metastatic tissue relative to normal colon, with a positive correlation between Nrf2 expression in matched primary and metastatic samples. In vitro experiments in CRC cell lines revealed that Nrf2 siRNA and brusatol, which is known to inhibit Nrf2, decreased viability and sensitised cells to irinotecan toxicity. Furthermore, brusatol effectively abrogated CRC tumour growth in subcutaneously and orthotopically-allografted mice, resulting in an average 8-fold reduction in luminescence at the study end-point (p=0.02). Our results highlight Nrf2 as a promising drug target in the treatment of CRC.
Collapse
Affiliation(s)
- Jonathan P Evans
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Boleslaw K Winiarski
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Paul A Sutton
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Robert P Jones
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Lorenzo Ressel
- Department of Veterinary Pathology, Institute of Veterinary Science, University of Liverpool, Liverpool, United Kingdom
| | - Carrie A Duckworth
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - D Mark Pritchard
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Vicky L Fretwell
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Elizabeth M Tweedle
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Christopher E Goldring
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Ian M Copple
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Daniel H Palmer
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom.,Clatterbridge Cancer Centre, Liverpool, United Kingdom
| | - Neil R Kitteringham
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
19
|
Wang L, Chan CEL, Wong ALA, Wong FC, Lim SW, Chinnathambi A, Alharbi SA, Lee LSU, Soo R, Yong WP, Lee SC, Ho PCL, Sethi G, Goh BC. Combined use of irinotecan with histone deacetylase inhibitor belinostat could cause severe toxicity by inhibiting SN-38 glucuronidation via UGT1A1. Oncotarget 2018; 8:41572-41581. [PMID: 28157715 PMCID: PMC5522258 DOI: 10.18632/oncotarget.15017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/07/2017] [Indexed: 12/27/2022] Open
Abstract
SN-38, the active metabolite of irinotecan, and histone deacetylase inhibitors (HDACis) such as belinostat, vorinostat and panobinostat, have all been shown to be deactivated by glucuronidation via UGTs. Since they all compete for UGTs for deactivation, we aimed to investigate the inhibitory effect of various HDACis on the glucuronidation of SN-38. This inhibitory effect was determined by measuring the formation rate of SN-38 glucuronide after SN-38 incubation with human recombinant UGT1A isoforms (1A1, 1A6, 1A7 and 1A9) and pooled human liver microsomes (HLM, wild type, UGT1A1*1*28 and UGT1A1*28*28 allelic variants), with and without HDACis. The data showed that belinostat at 100 and 200 µmol/L inhibited SN-38 glucuronidation via UGT1A1 in a dose-dependent manner, causing significant decrease in Vmax and CLint (p < 0.05) from 12.60 to 1.95 pmol/min/mg and 21.59 to 4.20 μL/min/mg protein respectively. Similarly, in HLMs, Vmax dropped from 41.13 to 10.54, 24.96 to 3.77 and 6.23 to 3.30 pmol/min/mg, and CLint reduced from 81.25 to 26.11, 29.22 to 6.10 and 5.40 to 1.34 µL/min/mg protein for the respective wild type, heterozygous and homozygous variants. Interestingly, belinostat at 200 µmol/L that is roughly equivalent to the average Cmax, 183 µmol/L of belinostat at a dose of 1,400 mg/m2 given intravenously once per day on days 1 to 5 every 3 weeks, was able to inhibit both heterozygous and homozygous variants to same extents (~64%). This highlights the potential clinical significance, as a large proportion of patients could be at risk of developing severe toxicity if irinotecan is co-administered with belinostat.
Collapse
Affiliation(s)
- Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, National University Health System, Singapore
| | - Chong En Linus Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacy, National University of Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Fang Cheng Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Siew Woon Lim
- Department of Pharmacy, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | | | - Ross Soo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Wei Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Soo Chin Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, National University Health System, Singapore.,Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth WA, Australia
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, National University Health System, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore
| |
Collapse
|
20
|
Li J, Bullara D, Du X, He H, Sofou S, Kevrekidis IG, Epstein IR, Xu B. Kinetic Analysis of Nanostructures Formed by Enzyme-Instructed Intracellular Assemblies against Cancer Cells. ACS NANO 2018; 12. [PMID: 29537820 PMCID: PMC5916050 DOI: 10.1021/acsnano.8b01016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Recent studies have demonstrated that enzyme-instructed self-assembly (EISA) in extra- or intracellular environments can serve as a multistep process for controlling cell fate. There is little knowledge, however, about the kinetics of EISA in the complex environments in or around cells. Here, we design and synthesize three dipeptidic precursors (ld-1-SO3, dl-1-SO3, dd-1-SO3), consisting of diphenylalanine (l-Phe-d-Phe, d-Phe-l-Phe, d-Phe-d-Phe, respectively) as the backbone, which are capped by 2-(naphthalen-2-yl)acetic acid at the N-terminal and by 2-(4-(2-aminoethoxy)-4-oxobutanamido)ethane-1-sulfonic acid at the C-terminal. On hydrolysis by carboxylesterases (CES), these precursors result in hydrogelators, which self-assemble in water at different rates. Whereas all three precursors selectively kill cancer cells, especially high-grade serous ovarian carcinoma cells, by undergoing intracellular EISA, dl-1-SO3 and dd-1-SO3 exhibit the lowest and the highest activities, respectively, against the cancer cells. This trend inversely correlates with the rates of converting the precursors to the hydrogelators in phosphate-buffered saline. Because CES exists both extra- and intracellularly, we use kinetic modeling to analyze the kinetics of EISA inside cells and to calculate the cytotoxicity of each precursor for killing cancer cells. Our results indicate that (i) the stereochemistry of the precursors affects the morphology of the nanostructures formed by the hydrogelators, as well as the rate of enzymatic conversion; (ii) decreased extracellular hydrolysis of precursors favors intracellular EISA inside the cells; (iii) the inherent features ( e.g., self-assembling ability and morphology) of the EISA molecules largely dictate the cytotoxicity of intracellular EISA. As the kinetic analysis of intracellular EISA, this work elucidates how the stereochemistry modulates EISA in the complex extra- and/or intracellular environment for developing anticancer molecular processes. Moreover, it provides insights for understanding the kinetics and cytotoxicity of aggregates of aberrant proteins or peptides formed inside and outside cells.
Collapse
Affiliation(s)
- Jie Li
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Domenico Bullara
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Xuewen Du
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
| | - Stavroula Sofou
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ioannis G. Kevrekidis
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Irving R. Epstein
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
- Corresponding Authors: ,
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA 02454, USA
- Corresponding Authors: ,
| |
Collapse
|
21
|
Cai L, Tang X, Guo L, An Y, Wang Y, Zheng J. Decreased Serum Levels of Carboxylesterase-2 in Patients with Ovarian Cancer. TUMORI JOURNAL 2018; 95:473-8. [DOI: 10.1177/030089160909500411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Carboxylesterase-2 has been identified as the key enzyme in the metabolic activation of irinotecan, a topoisomerase I inhibitor commonly used in the treatment of many solid tumors. Previous studies have shown that carboxylesterase-2 is down-regulated in colorectal cancer following progression of the disease. However, very limited information is available on carboxylesterase-2 expression in ovarian cancer. The aim of the present study was to detect the serum level and the tissue expression of carboxylesterase-2 in human ovarian cancer patients at different stages of the disease. Methods Carboxylesterase-2 levels in the serum of ovarian cancer patients were investigated by western blot and ELISA and in the tumor mass of ovarian cancer patients by western blot. Results Both the serum carboxylesterase-2 level and the expression of carboxylesterase-2 in tumor tissues were significantly different among patients at different stages of the disease (n = 40). No positive correlation was found between the serum carboxylesterase-2 level and the cancer antigen 125 level (n = 40). Serum carboxylesterase-2 is more sensitive than cancer antigen 125 in detecting the early stage patient with ovarian cancer. Conclusions Our results indicate that serum carboxylesterase-2 level might be a potential marker in the diagnosis of the early stage ovarian cancer.
Collapse
Affiliation(s)
- Liying Cai
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Xiaobo Tang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Lei Guo
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Yuan An
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Yuguang Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Jianhua Zheng
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
22
|
Lévi F, Karaboué A, Etienne-Grimaldi MC, Paintaud G, Focan C, Innominato P, Bouchahda M, Milano G, Chatelut E. Pharmacokinetics of Irinotecan, Oxaliplatin and 5-Fluorouracil During Hepatic Artery Chronomodulated Infusion: A Translational European OPTILIV Study. Clin Pharmacokinet 2017; 56:165-177. [PMID: 27393140 DOI: 10.1007/s40262-016-0431-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The combination of hepatic artery infusion (HAI) of irinotecan, 5-fluorouracil and oxaliplatin with intravenous cetuximab has safely achieved prolonged survival in colorectal cancer patients with extensive liver metastases and prior treatment. Systemic exposure to the drugs or their main metabolites was determined during the first course of chronomodulated triplet HAI in 11 patients and related to toxicities after one or three courses. Consistent trends were found between the area under the plasma concentration-time curve (AUC) values of irinotecan, 7-ethyl-10-hydroxycamptothecin (SN38; a bioactive metabolite), total oxaliplatin and platinum ultrafiltrate (P-UF), on the one hand, and subsequent leukopenia severity, on the other hand. Moreover, the maximum plasma concentration (C max) and the AUC of P-UF significantly predicted grades of diarrhoea (p = 0.004 and 0.017, respectively) and anaemia (p = 0.001 and 0.008, respectively) after the first course. Systemic drug exposure helps explain both the adverse events and the low rate of extrahepatic progression-a usual drawback of HAI chemotherapy-thus supporting upfront testing of the regimen. Systems optimization of chronomodulated HAI delivery could further reduce adverse events.
Collapse
Affiliation(s)
- Francis Lévi
- INSERM, UMRS 935, Team «Cancer Chronotherapy and Postoperative Liver Function», Campus CNRS, 7 rue Guy Môquet, Villejuif, France.
- Assistance Publique-Hopitaux de Paris, Medical Oncology Department, Hepatobiliary Centre, and Radiology Department, Paul Brousse Hospital, 14 avenue Paul Vaillant Couturier, Villejuif, France.
- Cancer Chronotherapy Unit, Warwick Medical School, University of Warwick, Gibbett Hill Road, Coventry, CV4 7AL, UK.
| | - Abdoulaye Karaboué
- INSERM, UMRS 935, Team «Cancer Chronotherapy and Postoperative Liver Function», Campus CNRS, 7 rue Guy Môquet, Villejuif, France
- AK-SCIENCE, Research and Therapeutic Innovation, Vitry-sur-Seine, France
| | | | - Gilles Paintaud
- Université François-Rabelais de Tours, CNRS, GICC UMR 7292, CHRU de Tours, Service de Pharmacologie-Toxicologie, Tours, France
| | - Christian Focan
- Department of Oncology, Centre Hospitalier Chrétien, Clinique Saint-Joseph, Liège, Belgium
| | - Pasquale Innominato
- INSERM, UMRS 935, Team «Cancer Chronotherapy and Postoperative Liver Function», Campus CNRS, 7 rue Guy Môquet, Villejuif, France
- Assistance Publique-Hopitaux de Paris, Medical Oncology Department, Hepatobiliary Centre, and Radiology Department, Paul Brousse Hospital, 14 avenue Paul Vaillant Couturier, Villejuif, France
- Cancer Chronotherapy Unit, Warwick Medical School, University of Warwick, Gibbett Hill Road, Coventry, CV4 7AL, UK
| | - Mohamed Bouchahda
- INSERM, UMRS 935, Team «Cancer Chronotherapy and Postoperative Liver Function», Campus CNRS, 7 rue Guy Môquet, Villejuif, France
- Assistance Publique-Hopitaux de Paris, Medical Oncology Department, Hepatobiliary Centre, and Radiology Department, Paul Brousse Hospital, 14 avenue Paul Vaillant Couturier, Villejuif, France
| | - Gérard Milano
- Laboratory of Oncopharmacology, Antoine Lacassagne Centre, Nice, France
| | - Etienne Chatelut
- Institut Claudius-Regaud, CRCT, Université de Toulouse, Inserm, UPS, Toulouse, France
| |
Collapse
|
23
|
Yao Y, Zhang P, Wang J, Chen J, Wang Y, Huang Y, Zhang Z, Xu F. Dissecting Target Toxic Tissue and Tissue Specific Responses of Irinotecan in Rats Using Metabolomics Approach. Front Pharmacol 2017; 8:122. [PMID: 28344557 PMCID: PMC5344918 DOI: 10.3389/fphar.2017.00122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 02/27/2017] [Indexed: 12/26/2022] Open
Abstract
As an anticancer agent, irinotecan (CPT-11) has been widely applied in clinical, especially in the treatment of colorectal cancer. However, its clinical use has long been limited by the side effects and potential tissue toxicity. To discriminate the target toxic tissues and dissect the specific response of target tissues after CPT-11 administration in rats, untargeted metabolomic study was conducted. First, differential metabolites between CPT-11 treated group and control group in each tissue were screened out. Then, based on fold changes of these differential metabolites, principal component analysis and hierarchical cluster analysis were performed to visualize the degree and specificity of the influences of CPT-11 on the metabolic profiles of nine tissues. Using this step-wise method, ileum, jejunum, and liver were finally recognized as target toxic tissues. Furthermore, tissue specific responses of liver, ileum, and jejunum to CPT-11 were dissected and specific differential metabolites were screened out. Perturbations in Krebs cycle, amino acid, purine and bile acid metabolism were observed in target toxic tissues. In conclusion, our study put forward a new approach to dissect target toxic tissues and tissue specific responses of CPT-11 using metabolomics.
Collapse
Affiliation(s)
- Yiran Yao
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Jing Wang
- School of Pharmacy, Shanxi University of Chinese Medicine Xianyang, China
| | - Jiaqing Chen
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Yong Wang
- Jiangsu Institute for Food and Drug Control Nanjing, China
| | - Yin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical UniversityNanjing, China; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical UniversityNanjing, China; State Key Laboratory of Natural Medicine, China Pharmaceutical UniversityNanjing, China
| |
Collapse
|
24
|
Coriat R, Faivre SJ, Mir O, Dreyer C, Ropert S, Bouattour M, Desjardins R, Goldwasser F, Raymond E. Pharmacokinetics and safety of DTS-108, a human oligopeptide bound to SN-38 with an esterase-sensitive cross-linker in patients with advanced malignancies: a Phase I study. Int J Nanomedicine 2016; 11:6207-6216. [PMID: 27920527 PMCID: PMC5123727 DOI: 10.2147/ijn.s110274] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND DTS-108 is a hydrosoluble prodrug, where the SN-38 moiety is covalently linked to a 20-amino acid vector peptide by a specific esterase-sensitive cross-linker, releasing 7-ethyl-10-hydroxycampthotecin (SN-38) by esterase bond cleavage. METHODS The pharmacokinetics of DTS-108, adverse events graded according to NCI-CTCv3.1, dose-limiting toxicities at cycle 1, the maximum tolerated dose (MTD), and the recommended Phase II dose (RP2D) of intravenous DTS-108 (1-2 hours) every 2 weeks were evaluated in a first-in-human Phase I study in patients with advanced/metastatic carcinomas, according to an accelerated dose escalation design. SN-38 and SN-38 glucuronide (SN-38G) levels were evaluated with fluorescence high-performance liquid chromatography (HPLC) test, then liquid chromatography-tandem mass spectrometry (LC/MS/MS) methods. RESULTS Forty-two patients received DTS-108 across 14 dosing cohorts (range 3-416 mg/m2). At 416 mg/m2, three out of six patients had grade 4 neutropenia thereby defining the MTD and the RP2D at 313 mg/m2. Fluorescence HPLC was inaccurate to quantify DTS-108 and its metabolites (SN-38 and SN-38G). New processes and analytical LC/MS/MS methods for testing SN-38 were implemented. At a dose of 313 mg/m2, mean DTS-108, SN-38, and SN-38G area under the plasma concentration-time curve to infinity (coefficients of variation %) were 439,293 (24%), 1,992 (34%), and 4,538 (46%) h·ng/mL. Stable disease (according to Response Evaluation Criteria in Solid Tumors) was observed in nine patients. CONCLUSION Assessing SN-38 concentration using fluorescence HPLC is questionable since this method failed to monitor dose escalation of DTS-108, a new topoisomerase I inhibitor, due to ex vivo degradation. LC/MS/MS methods were consistent in evaluating SN-38 exposures allowing drug monitoring. The maximum tolerated dose of DTS-108 was 416 mg/m2. The RP2D for intravenous DTS-108 was 313 mg/m2 every 2 weeks in patients with advanced/metastatic solid tumors.
Collapse
Affiliation(s)
- Romain Coriat
- Gastroenterology and Digestive Oncology Unit, Cochin Teaching Hospital, Université Paris Descartes Sorbonne Paris Cité, Paris
| | - Sandrine J Faivre
- Department of Medical Oncology, Beaujon Teaching Hospital, Université Paris Diderot, Paris 7, Clichy
| | - Olivier Mir
- Department of Medical Oncology, Cochin Teaching Hospital, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Chantal Dreyer
- Department of Medical Oncology, Beaujon Teaching Hospital, Université Paris Diderot, Paris 7, Clichy
| | - Stanislas Ropert
- Department of Medical Oncology, Cochin Teaching Hospital, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Mohammed Bouattour
- Department of Medical Oncology, Beaujon Teaching Hospital, Université Paris Diderot, Paris 7, Clichy
| | | | - François Goldwasser
- Department of Medical Oncology, Cochin Teaching Hospital, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Eric Raymond
- Groupe Hospitalier Paris Saint-Joseph, Paris, France
| |
Collapse
|
25
|
Kühl AA, Erben U, Cieluch C, Spieckermann S, Gröne J, Lohneis P, Pape UF, Arsenic R, Utku N. Tissue-infiltrating plasma cells are an important source of carboxylesterase 2 contributing to the therapeutic efficacy of prodrugs. Cancer Lett 2016; 378:51-8. [DOI: 10.1016/j.canlet.2016.04.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 12/16/2022]
|
26
|
Bala V, Rao S, Prestidge CA. Facilitating gastrointestinal solubilisation and enhanced oral absorption of SN38 using a molecularly complexed silica-lipid hybrid delivery system. Eur J Pharm Biopharm 2016; 105:32-9. [DOI: 10.1016/j.ejpb.2016.05.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/26/2016] [Accepted: 05/26/2016] [Indexed: 01/16/2023]
|
27
|
Kang MH, Wang J, Makena MR, Lee JS, Paz N, Hall CP, Song MM, Calderon RI, Cruz RE, Hindle A, Ko W, Fitzgerald JB, Drummond DC, Triche TJ, Reynolds CP. Activity of MM-398, nanoliposomal irinotecan (nal-IRI), in Ewing's family tumor xenografts is associated with high exposure of tumor to drug and high SLFN11 expression. Clin Cancer Res 2016; 21:1139-50. [PMID: 25733708 DOI: 10.1158/1078-0432.ccr-14-1882] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE To determine the pharmacokinetics and the antitumor activity in pediatric cancer models of MM-398, a nanoliposomal irinotecan (nal-IRI). EXPERIMENTAL DESIGN Mouse plasma and tissue pharmacokinetics of nal-IRI and the current clinical formulation of irinotecan were characterized. In vivo activity of irinotecan and nal-IRI was compared in xenograft models (3 each in nu/nu mice) of Ewing's sarcoma family of tumors (EFT), neuroblastoma (NB), and rhabdomyosarcoma (RMS). SLFN11 expression was assessed by Affymetrix HuEx arrays, Taqman RT-PCR, and immunoblotting. RESULTS Plasma and tumor concentrations of irinotecan and SN-38 (active metabolite) were approximately 10-fold higher for nal-IRI than for irinotecan. Two doses of NAL-IRI (10 mg/kg/dose) achieved complete responses maintained for >100 days in 24 of 27 EFT-xenografted mice. Event-free survival for mice with RMS and NB was significantly shorter than for EFT. High SLFN11 expression has been reported to correlate with sensitivity to DNA damaging agents; median SLFN11 mRNA expression was >100-fold greater in both EFT cell lines and primary tumors compared with NB or RMS cell lines or primary tumors. Cytotoxicity of SN-38 inversely correlated with SLFN11 mRNA expression in 20 EFT cell lines. CONCLUSIONS In pediatric solid tumor xenografts, nal-IRI demonstrated higher systemic and tumor exposures to SN-38 and improved antitumor activity compared with the current clinical formulation of irinotecan. Clinical studies of nal-IRI in pediatric solid tumors (especially EFT) and correlative studies to determine if SLFN11 expression can serve as a biomarker to predict nal-IRI clinical activity are warranted.
Collapse
Affiliation(s)
- Min H Kang
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Jing Wang
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Monish R Makena
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Joo-Sang Lee
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Nancy Paz
- Merrimack Pharmaceuticals, Cambridge, Massachusetts
| | - Connor P Hall
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Michael M Song
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Ruben I Calderon
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Riza E Cruz
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Ashly Hindle
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Winford Ko
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| | | | | | - Timothy J Triche
- Department of Pathology Keck School of Medicine, University of Southern California, Los Angeles, California
| | - C Patrick Reynolds
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas. Departments of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas.
| |
Collapse
|
28
|
Nakatsuji M, Inoue H, Kohno M, Saito M, Tsuge S, Shimizu S, Ishida A, Ishibashi O, Inui T. Human Lipocalin-Type Prostaglandin D Synthase-Based Drug Delivery System for Poorly Water-Soluble Anti-Cancer Drug SN-38. PLoS One 2015; 10:e0142206. [PMID: 26529243 PMCID: PMC4631600 DOI: 10.1371/journal.pone.0142206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/19/2015] [Indexed: 01/18/2023] Open
Abstract
Lipocalin-type prostaglandin D synthase (L-PGDS) is a member of the lipocalin superfamily, which is composed of secretory transporter proteins, and binds a wide variety of small hydrophobic molecules. Using this function, we have reported the feasibility of using L-PGDS as a novel drug delivery vehicle for poorly water-soluble drugs. In this study, we show the development of a drug delivery system using L-PGDS, one that enables the direct clinical use of 7-ethyl-10-hydroxy-camptothecin (SN-38), a poorly water-soluble anti-cancer drug. In the presence of 2 mM L-PGDS, the concentration of SN-38 in PBS increased 1,130-fold as compared with that in PBS. Calorimetric experiments revealed that L-PGDS bound SN-38 at a molecular ratio of 1:3 with a dissociation constant value of 60 μM. The results of an in vitro growth inhibition assay revealed that the SN-38/L-PGDS complexes showed high anti-tumor activity against 3 human cancer cell lines, i.e., Colo201, MDA-MB-231, and PC-3 with a potency similar to that of SN-38 used alone. The intravenous administration of SN-38/L-PGDS complexes to mice bearing Colo201 tumors showed a pronounced anti-tumor effect. Intestinal mucositis, which is one of the side effects of this drug, was not observed in mice administered SN-38/L-PGDS complexes. Taken together, L-PGDS enables the direct usage of SN-38 with reduced side effects.
Collapse
Affiliation(s)
- Masatoshi Nakatsuji
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Haruka Inoue
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Masaki Kohno
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Mayu Saito
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Syogo Tsuge
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Shota Shimizu
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Atsuko Ishida
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Osamu Ishibashi
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Takashi Inui
- Department of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- * E-mail:
| |
Collapse
|
29
|
Li J, Kuang Y, Shi J, Zhou J, Medina JE, Zhou R, Yuan D, Yang C, Wang H, Yang Z, Liu J, Dinulescu DM, Xu B. Enzyme-Instructed Intracellular Molecular Self-Assembly to Boost Activity of Cisplatin against Drug-Resistant Ovarian Cancer Cells. Angew Chem Int Ed Engl 2015; 54:13307-11. [PMID: 26365295 PMCID: PMC4681442 DOI: 10.1002/anie.201507157] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Indexed: 01/29/2023]
Abstract
Anticancer drug resistance demands innovative approaches that boost the activity of drugs against drug-resistant cancers without increasing the systemic toxicity. Here we show the use of enzyme-instructed self-assembly (EISA) to generate intracellular supramolecular assemblies that drastically boost the activity of cisplatin against drug-resistant ovarian cancer cells. We design and synthesize small peptide precursors as the substrates of carboxylesterase (CES). CES cleaves the ester bond pre-installed on the precursors to form the peptides that self-assemble in water to form nanofibers. At the optimal concentrations, the precursors themselves are innocuous to cells, but they double or triple the activity of cisplatin against the drug-resistant ovarian cancer cells. This work illustrates a simple, yet fundamental, new way to introduce non-cytotoxic components into combination therapies with cisplatin without increasing the systemic burden or side effects.
Collapse
Affiliation(s)
- Jie Li
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Yi Kuang
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Junfeng Shi
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Jie Zhou
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Jamie E Medina
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 (USA)
| | - Rong Zhou
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Dan Yuan
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Cuihong Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192 (P.R. China)
| | - Huaimin Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071 (China)
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071 (China)
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192 (P.R. China)
| | - Daniela M Dinulescu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 (USA).
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA).
| |
Collapse
|
30
|
Li J, Kuang Y, Shi J, Zhou J, Medina JE, Zhou R, Yuan D, Yang C, Wang H, Yang Z, Liu J, Dinulescu DM, Xu B. Enzyme‐Instructed Intracellular Molecular Self‐Assembly to Boost Activity of Cisplatin against Drug‐Resistant Ovarian Cancer Cells. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201507157] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Jie Li
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Yi Kuang
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Junfeng Shi
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Jie Zhou
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Jamie E. Medina
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 (USA)
| | - Rong Zhou
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Dan Yuan
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| | - Cuihong Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192 (P.R. China)
| | - Huaimin Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071 (China)
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300071 (China)
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300192 (P.R. China)
| | - Daniela M. Dinulescu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115 (USA)
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South St, Waltham, MA 02454 (USA)
| |
Collapse
|
31
|
Nipp RD, Ryan DP. Predicting a response to FOLFIRINOX in pancreatic cancer. J Natl Cancer Inst 2015; 107:djv175. [PMID: 26025325 DOI: 10.1093/jnci/djv175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ryan D Nipp
- Massachusetts General Hospital Cancer Center
| | | |
Collapse
|
32
|
Jakobsen AK, Lauridsen KL, Samuel EB, Proszek J, Knudsen BR, Hager H, Stougaard M. Correlation between topoisomerase I and tyrosyl-DNA phosphodiesterase 1 activities in non-small cell lung cancer tissue. Exp Mol Pathol 2015; 99:56-64. [PMID: 25987486 DOI: 10.1016/j.yexmp.2015.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/04/2015] [Accepted: 05/14/2015] [Indexed: 12/29/2022]
Abstract
Topoisomerase I (TOP1) regulates DNA topology during replication and transcription whereas tyrosyl-DNA phosphodiesterase 1 (TDP1) is involved in the repair of several types of DNA damages, including damages from defective TOP1 catalysis. TOP1 is the target of chemotherapeutic drugs of the camptothecin family (CPT). TDP1 has in cell line based assays been shown to counteract the effect of CPT. We have quantified the enzymatic activities of TOP1 and TDP1 in paired (tumor and adjacent non-tumor) samples from non-small cell lung cancer (NSCLC) patients and show that in NSCLC TOP1 and TDP1 activities are significantly upregulated in the tumor tissue. Furthermore, we found a positive correlation between the TDP1 activity and the tumor percentage (TOP1 activity did not correlate with the tumor percentage) as well as between the activities of TOP1 and TDP1 both within the tumor and the non-tumor group. That TDP1 activity was upregulated in all tumor samples and correlated with the tumor percentage suggest that it must play a highly important function in NSCLC. This could be to protect against TOP1 mediated DNA damage as the activity of TOP1 likewise was upregulated in the majority of tumor samples and correlated positively to the TDP1 activity. Regardless, the finding that the TOP1 and TDP1 activities are upregulated and correlate positively suggests that combinatorial treatment targeting both activities could be advantageous in NSCLC.
Collapse
Affiliation(s)
| | | | | | - Joanna Proszek
- Department of Pathology, Aarhus University Hospital, Denmark
| | - Birgitta Ruth Knudsen
- Department of Molecular Biology and Genetics, Aarhus University, Denmark; Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Denmark
| | - Henrik Hager
- Department of Pathology, Aarhus University Hospital, Denmark; Department of Clinical Pathology, Vejle Hospital, Denmark
| | | |
Collapse
|
33
|
Papaevangelou E, Almeida GS, Jamin Y, Robinson SP, deSouza NM. Diffusion-weighted MRI for imaging cell death after cytotoxic or apoptosis-inducing therapy. Br J Cancer 2015; 112:1471-9. [PMID: 25880014 PMCID: PMC4453679 DOI: 10.1038/bjc.2015.134] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/10/2015] [Accepted: 03/17/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Non-invasive serial imaging is desirable to detect processes such as necrotic and apoptotic cell death in cancer patients undergoing treatment. This study investigated the use of diffusion-weighted (DW-) magnetic resonance imaging (MRI) for imaging cell death induced by either a cytotoxic drug (irinotecan), or the apoptosis-inducing agent birinapant, in human tumour xenografts in vivo. METHODS Nude mice bearing human SW620 colon carcinoma xenografts were treated with vehicle, irinotecan (50 mg kg(-1)) or birinapant (30 mg kg(-1)) for up to 5 days. DW-MRI was performed prior to and on days 1, 3 and 5 during treatment. Assessment of tumour apoptosis and necrosis ex vivo was used to validate the imaging findings. RESULTS Both irinotecan and birinapant induced significant tumour growth delay. Irinotecan induced a small increase in the tumour apparent diffusion coefficient (ADC) after 1 day, with a 20 and 30% increase at days 3 and 5 respectively. ADC was unchanged in the vehicle- and birinapant-treated tumours despite a growth delay in the latter. Histological analysis showed that irinotecan increased necrosis at days 3 and 5, and induced apoptosis after 1 day, compared with vehicle. Birinapant induced apoptosis after day 3, but had no effect on tumour necrosis. CONCLUSIONS Tumour ADC changes after irinotecan treatment were associated with the induction of a mixture of necrotic and apoptotic cell death, whereas induction of apoptosis alone with birinapant was not sufficient to induce changes in tissue microstructure that were detectable with DW-MRI. ADC is a useful non-invasive biomarker for early detection of response to cytotoxic drugs, but false negatives may arise while detecting apoptotic response to birinapant.
Collapse
Affiliation(s)
- E Papaevangelou
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - G S Almeida
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - Y Jamin
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - S P Robinson
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| | - N M deSouza
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, 15 Cotswold Road, Belmont, Sutton, Surrey SM2 5NG, UK
| |
Collapse
|
34
|
Neijzen R, Wong MQ, Gill N, Wang H, Karim T, Anantha M, Strutt D, Waterhouse D, Bally MB, Tai IT, Ng SS, Yapp DT. Irinophore C™, a lipid nanoparticulate formulation of irinotecan, improves vascular function, increases the delivery of sequentially administered 5-FU in HT-29 tumors, and controls tumor growth in patient derived xenografts of colon cancer. J Control Release 2015; 199:72-83. [DOI: 10.1016/j.jconrel.2014.11.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/18/2014] [Accepted: 11/26/2014] [Indexed: 11/29/2022]
|
35
|
In Vivo Evaluation of Irinotecan-Loaded QuadraSphere Microspheres for Use in Chemoembolization of VX2 Liver Tumors. J Vasc Interv Radiol 2014; 25:1727-35.e1. [DOI: 10.1016/j.jvir.2014.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 01/01/2023] Open
|
36
|
Molecular mechanisms of fenofibrate-induced metabolic catastrophe and glioblastoma cell death. Mol Cell Biol 2014; 35:182-98. [PMID: 25332241 DOI: 10.1128/mcb.00562-14] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Fenofibrate (FF) is a common lipid-lowering drug and a potent agonist of the peroxisome proliferator-activated receptor alpha (PPARα). FF and several other agonists of PPARα have interesting anticancer properties, and our recent studies demonstrate that FF is very effective against tumor cells of neuroectodermal origin. In spite of these promising anticancer effects, the molecular mechanism(s) of FF-induced tumor cell toxicity remains to be elucidated. Here we report a novel PPARα-independent mechanism explaining FF's cytotoxicity in vitro and in an intracranial mouse model of glioblastoma. The mechanism involves accumulation of FF in the mitochondrial fraction, followed by immediate impairment of mitochondrial respiration at the level of complex I of the electron transport chain. This mitochondrial action sensitizes tested glioblastoma cells to the PPARα-dependent metabolic switch from glycolysis to fatty acid β-oxidation. As a consequence, prolonged exposure to FF depletes intracellular ATP, activates the AMP-activated protein kinase-mammalian target of rapamycin-autophagy pathway, and results in extensive tumor cell death. Interestingly, autophagy activators attenuate and autophagy inhibitors enhance FF-induced glioblastoma cytotoxicity. Our results explain the molecular basis of FF-induced glioblastoma cytotoxicity and reveal a new supplemental therapeutic approach in which intracranial infusion of FF could selectively trigger metabolic catastrophe in glioblastoma cells.
Collapse
|
37
|
Distribution and quantification of irinotecan and its active metabolite SN-38 in colon cancer murine model systems using MALDI MSI. Anal Bioanal Chem 2014; 407:2107-16. [PMID: 25311193 DOI: 10.1007/s00216-014-8237-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 09/24/2014] [Accepted: 10/02/2014] [Indexed: 10/24/2022]
Abstract
Tissue distribution and quantitative analysis of small molecules is a key to assess the mechanism of drug action and evaluate treatment efficacy. The prodrug irinotecan (CPT-11) is widely used for chemotherapeutic treatment of colorectal cancer. CPT-11 requires conversion into its active metabolite SN-38 to exert the desired pharmacological effect. MALDI-Fourier transform ion cyclotron resonance (FT-ICR) and MALDI-time-of-flight (TOF) mass spectrometry imaging (MSI) were performed for detection of CPT-11 and SN-38 in tissue sections from mice post CPT-11 injection. In-depth information was gained about the distribution and quantity of drug compounds in normal and tumor tissue. The prodrug was metabolized, as proven by the detection of SN-38 in liver, kidney and digestive tract. In tumors from genetic mouse models for colorectal cancer (Apc (1638N/wt) x pvillin-Kras (V12G) ), CPT-11 was detected but not the active metabolite. In order to correlate drug distribution relative to vascularization, MALDI data were superimposed with CD31 (PECAM-1) immunohistochemistry. This analysis indicated that intratumoral access of CPT-11 mainly occurred by extravasation from microvessels. The present study exploits the power of MALDI MSI in drug analysis, and presents a novel approach to monitor drug distribution in relation to vessel functionality in preclinical and clinical research.
Collapse
|
38
|
Gene expression differences in primary colorectal tumors and matched liver metastases: chemotherapy related or tumoral heterogeneity? Clin Transl Oncol 2014; 17:322-9. [PMID: 25301403 DOI: 10.1007/s12094-014-1233-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/20/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Treatment of metastatic colorectal cancer (mCRC) is generally based on genetic testing performed in primary tumor biopsies, but whether the genomic status of primary tumors is identical to that of metastases is not well known. We compared the gene expression profiles of formalin-fixed paraffin-embedded (FFPE) biopsies of colorectal primary tumors and matched liver metastases. PATIENTS AND METHODS We compared the expression of 18 genes in FFPE CRC tumors and their matched liver metastases from 32 patients. The expression of each gene in CRC primary tumors and their matched liver metastases was tested using Student's t test for paired samples. Pairwise correlations of each gene in the primary tumors and matched liver metastases were evaluated by Pearson's correlation coefficient. RESULTS The expression of six genes was significantly different in primary tumors compared with their matched liver metastases [CXCR4 (p < 0.001), THBS1 (p = 0.007), MMP 9 (p = 0.048), GST Pi (p = 0.050), TYMP (p = 0.042) and DPYD (p < 0.001)]. For the remaining genes, where no significant differences were observed, only SMAD4 (r s = 0.447, p = 0.010), ERCC1 (r s = 0.423, p = 0.016) and VEGF A (r s = 0.453, p = 0.009) showed significant correlation in expression between the two tissues. Therefore, we only detected similar gene expression levels between the tumor and the metastases in these three markers. CONCLUSIONS We only found similar gene expression levels between the tumor and the metastases in three genes (SMAD4, ERCC1, and VEGF A). However, our study could not assess whether the differences in gene expression were secondary to tumoral heterogeneity or to molecular changes induced by previous chemotherapy.
Collapse
|
39
|
Jensen PR, Serra SC, Miragoli L, Karlsson M, Cabella C, Poggi L, Venturi L, Tedoldi F, Lerche MH. Hyperpolarized [1,3-13C2]ethyl acetoacetate is a novel diagnostic metabolic marker of liver cancer. Int J Cancer 2014; 136:E117-26. [DOI: 10.1002/ijc.29162] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 06/20/2014] [Accepted: 07/23/2014] [Indexed: 12/17/2022]
Affiliation(s)
| | | | - Luigi Miragoli
- Centro Ricerche Bracco; Bracco Imaging Spa; Colleretto Giacosa (TO) Italy
| | | | - Claudia Cabella
- Centro Ricerche Bracco; Bracco Imaging Spa; Colleretto Giacosa (TO) Italy
| | - Luisa Poggi
- Centro Ricerche Bracco; Bracco Imaging Spa; Colleretto Giacosa (TO) Italy
| | - Luca Venturi
- Center of Preclinical Imaging; University of Torino (Italy); Colleretto Giacosa (TO) Italy
| | - Fabio Tedoldi
- Centro Ricerche Bracco; Bracco Imaging Spa; Colleretto Giacosa (TO) Italy
| | | |
Collapse
|
40
|
Liu H, Lu H, Liao L, Zhang X, Gong T, Zhang Z. Lipid nanoparticles loaded with 7-ethyl-10-hydroxycamptothecin-phospholipid complex: in vitro and in vivo studies. Drug Deliv 2014; 22:701-9. [PMID: 24625262 DOI: 10.3109/10717544.2014.895069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
7-Ethyl-10-hydroxy-camptothecin (SN38), the active metabolite of irinotecan (CPT-11), is an effective antineoplastic agent against many malignancies. Although it is 100-fold to 1000-fold more potent than CPT-11, the clinical utility of SN38 has been extremely restricted because of its poor solubility in any pharmaceutically acceptable solvents. The aim of this study was to develop SN38 nanoparticles (SN38-PC-LNs) using pharmaceutically acceptable excipients, and investigate the therapeutic efficacies in vitro and in vivo. SN38-phospholipid complex (SN38-PC) was prepared and loaded into lipid nanoparticles. The particle size was approximately 200 nm with a narrow size distribution. A high encapsulation efficiency of 88.11% ± 1.41% and drug loading of 9.55% ± 0.84% were achieved under the optimal condition. SN38-PC-LNs exhibited potent cytotoxic effects against a panel of human tumor cell lines (HT-29, HepG2, A549 and MCF-7). In vivo evaluation proved the enhanced antitumor efficacy of SN38-PC-LNs in mice bearing S180 tumor as well. The results from this study demonstrated an effective formulation of SN38 has been developed, which is promising for the delivery of SN38 for tumor chemotherapy.
Collapse
Affiliation(s)
- Huan Liu
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education , Sichuan University , Sichuan , People's Republic of China
| | - Hua Lu
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education , Sichuan University , Sichuan , People's Republic of China
| | - Longfei Liao
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education , Sichuan University , Sichuan , People's Republic of China
| | - Xuanmiao Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education , Sichuan University , Sichuan , People's Republic of China
| | - Tao Gong
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education , Sichuan University , Sichuan , People's Republic of China
| | - Zhirong Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education , Sichuan University , Sichuan , People's Republic of China
| |
Collapse
|
41
|
Noble CO, Krauze MT, Drummond DC, Forsayeth J, Hayes ME, Beyer J, Hadaczek P, Berger MS, Kirpotin DB, Bankiewicz KS, Park JW. Pharmacokinetics, tumor accumulation and antitumor activity of nanoliposomal irinotecan following systemic treatment of intracranial tumors. Nanomedicine (Lond) 2014; 9:2099-108. [PMID: 24494810 DOI: 10.2217/nnm.13.201] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
AIM We sought to evaluate nanoliposomal irinotecan as an intravenous treatment in an orthotopic brain tumor model. MATERIALS & METHODS Nanoliposomal irinotecan was administered intravenously in the intracranial U87MG brain tumor model in mice, and irinotecan and SN-38 levels were analyzed in malignant and normal tissues. Therapy studies were performed in comparison to free irinotecan and control treatments. RESULTS Tissue analysis demonstrated favorable properties for nanoliposomal irinotecan, including a 10.9-fold increase in tumor AUC for drug compared with free irinotecan and 35-fold selectivity for tumor versus normal tissue exposure. As a therapy for orthotopic brain tumors, nanoliposomal irinotecan showed a mean survival time of 54.2 versus 29.5 days for free irinotecan. A total of 33% of the animals receiving nanoliposomal irinotecan showed no residual tumor by study end compared with no survivors in the other groups. CONCLUSION Nanoliposomal irinotecan administered systemically provides significant pharmacologic advantages and may be an efficacious therapy for brain tumors.
Collapse
Affiliation(s)
- Charles O Noble
- Division of Hematology-Oncology, University of California San Francisco, San Francisco, CA 94115, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Stenvang J, Kümler I, Nygård SB, Smith DH, Nielsen D, Brünner N, Moreira JMA. Biomarker-guided repurposing of chemotherapeutic drugs for cancer therapy: a novel strategy in drug development. Front Oncol 2013; 3:313. [PMID: 24400218 PMCID: PMC3872326 DOI: 10.3389/fonc.2013.00313] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Accepted: 12/10/2013] [Indexed: 12/29/2022] Open
Abstract
Cancer is a leading cause of mortality worldwide and matters are only set to worsen as its incidence continues to rise. Traditional approaches to combat cancer include improved prevention, early diagnosis, optimized surgery, development of novel drugs, and honing regimens of existing anti-cancer drugs. Although discovery and development of novel and effective anti-cancer drugs is a major research area, it is well known that oncology drug development is a lengthy process, extremely costly and with high attrition rates. Furthermore, those drugs that do make it through the drug development mill are often quite expensive, laden with severe side-effects and unfortunately, to date, have only demonstrated minimal increases in overall survival. Therefore, a strong interest has emerged to identify approved non-cancer drugs that possess anti-cancer activity, thus shortcutting the development process. This research strategy is commonly known as drug repurposing or drug repositioning and provides a faster path to the clinics. We have developed and implemented a modification of the standard drug repurposing strategy that we review here; rather than investigating target-promiscuous non-cancer drugs for possible anti-cancer activity, we focus on the discovery of novel cancer indications for already approved chemotherapeutic anti-cancer drugs. Clinical implementation of this strategy is normally commenced at clinical phase II trials and includes pre-treated patients. As the response rates to any non-standard chemotherapeutic drug will be relatively low in such a patient cohort it is a pre-requisite that such testing is based on predictive biomarkers. This review describes our strategy of biomarker-guided repurposing of chemotherapeutic drugs for cancer therapy, taking the repurposing of topoisomerase I (Top1) inhibitors and Top1 as a potential predictive biomarker as case in point.
Collapse
Affiliation(s)
- Jan Stenvang
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, University of Copenhagen , Copenhagen , Denmark ; Danish Centre for Translational Breast Cancer Research , Copenhagen , Denmark
| | - Iben Kümler
- Department of Oncology, Center for Cancer Research, Herlev Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Sune Boris Nygård
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, University of Copenhagen , Copenhagen , Denmark
| | - David Hersi Smith
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, University of Copenhagen , Copenhagen , Denmark ; DAKO A/S , Glostrup , Denmark
| | - Dorte Nielsen
- Department of Oncology, Center for Cancer Research, Herlev Hospital, University of Copenhagen , Copenhagen , Denmark
| | - Nils Brünner
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, University of Copenhagen , Copenhagen , Denmark ; Danish Centre for Translational Breast Cancer Research , Copenhagen , Denmark
| | - José M A Moreira
- Faculty of Health and Medical Sciences, Department of Veterinary Disease Biology, Section for Molecular Disease Biology and Sino-Danish Breast Cancer Research Centre, University of Copenhagen , Copenhagen , Denmark ; Danish Centre for Translational Breast Cancer Research , Copenhagen , Denmark
| |
Collapse
|
43
|
Jones RP, Sutton P, Greensmith RMD, Santoyo-Castelazo A, Carr DF, Jenkins R, Rowe C, Hamlett J, Park BK, Terlizzo M, O'Grady E, Ghaneh P, Fenwick SW, Malik HZ, Poston GJ, Kitteringham NR. Hepatic activation of irinotecan predicts tumour response in patients with colorectal liver metastases treated with DEBIRI: exploratory findings from a phase II study. Cancer Chemother Pharmacol 2013; 72:359-68. [PMID: 23756919 DOI: 10.1007/s00280-013-2199-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/20/2013] [Indexed: 02/06/2023]
Abstract
PURPOSE The response of colorectal liver metastases to the cytotoxic agent irinotecan varies widely. Attempts to correlate tumour metabolism with response have been mixed. This study investigated the hepatic metabolism of irinotecan as a potential predictor of tumour response to irinotecan-eluting beads (DEBIRI). METHODS Ten patients with colorectal liver metastases were treated with 200 mg irinotecan (as DEBIRI) as part of the PARAGON II study. Hepatic expression of key metabolising enzymes was measured using mass spectrometry-based proteomics. Serum drug concentrations and hepatic irinotecan metabolism were characterised and correlated with tumour response. RESULTS Serum concentrations of irinotecan metabolites did not correlate with hepatic metabolism or pathological response. There was a strong correlation between hepatic CES-2 expression and activation of irinotecan (r (2) = 0.96, p < 0.001). Patients with a UGT1A1*28 6/7 SNP showed no difference in drug metabolism or pathological response. Hepatic CES-2 mediated activation of irinotecan clearly correlated with tumour replacement by fibrosis (r (2) = 0.54, p = 0.01). CONCLUSION This study provides the first evidence that hepatic activation of irinotecan predicts tumour response. Delivery of liver-targeted irinotecan to normal liver tissue rather than tumour may be a more rational approach to maximise response.
Collapse
Affiliation(s)
- R P Jones
- School of Cancer Studies, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hare JI, Neijzen RW, Anantha M, Dos Santos N, Harasym N, Webb MS, Allen TM, Bally MB, Waterhouse DN. Treatment of colorectal cancer using a combination of liposomal irinotecan (Irinophore C™) and 5-fluorouracil. PLoS One 2013; 8:e62349. [PMID: 23626804 PMCID: PMC3633892 DOI: 10.1371/journal.pone.0062349] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 03/22/2013] [Indexed: 01/09/2023] Open
Abstract
Purpose To investigate the use of liposomal irinotecan (Irinophore C™) plus or minus 5-fluorouracil (5-FU) for the treatment of colorectal cancer. Experimental Design The effect of irinotecan (IRI) and/or 5-FU exposure times on cytotoxicity was assessed in vitro against HT-29 or LS174T human colon carcinoma cells. The pharmacokinetics and biodistribution of Irinophore C™ (IrC™) and 5-FU, administered alone or in combination, were compared in vivo. A subcutaneous model of HT-29 human colorectal cancer in Rag2-M mice was utilized to assess the efficacy of IrC™ alone, and in combination with 5-FU. Results The cytotoxicity of IRI and 5-FU were strongly dependent on exposure time. Synergistic interactions were observed following prolonged exposure to IRI/5-FU combinations. Pharmacokinetics/biodistribution studies demonstrated that the 5-FU elimination rate was decreased significantly when 5-FU was co-administered intravenously with IrC™, versus alone. Significant decreases in 5-FU elimination were also observed in plasma, with an associated increase of 5-FU in some tissues when 5-FU was given by intraperitoneal injection and IrC™ was given intravenously. The elimination of IrC™ was not significantly different when administered alone or in combination with 5-FU. Therapeutic studies demonstrated that single agent IrC™ was significantly more effective than the combination of IRI/5-FU; surprisingly, IrC™/5-FU combinations were no more effective than IrC™ alone. The administration of combinations of 5-FU (16 mg/kg) and IrC™ (60 mg IRI/kg) showed increased toxicity when compared to IrC™ alone. Treatment with IrC™ alone (60 mg IRI/kg) delayed the time required for a 5-fold increase in initial tumor volume to day 49, compared to day 23 for controls. When IrC™ (40 mg IRI/kg) was used in combination with 5-FU (16 mg/kg), the time to increase tumor volume 5-fold was 43 days, which was comparable to that achieved when using IrC™ alone (40 mg IRI/kg). Conclusions Single agent IrC™ was well tolerated and has significant therapeutic potential. IrC™ may be a suitable replacement for IRI treatment, but its use with free 5-FU is complicated by IrC™-engendered changes in 5-FU pharmacokinetics/biodistribution which are associated with increased toxicity when using the combination.
Collapse
Affiliation(s)
- Jennifer I. Hare
- Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Robert W. Neijzen
- Department of Pharmaceutical Science, Universiteit Utrecht, Utrecht, Netherlands
| | - Malathi Anantha
- Experimental Therapeutics, BC Cancer Agency, Vancouver, Canada
| | | | | | - Murray S. Webb
- Centre for Drug Research and Development, Vancouver, Canada
| | - Theresa M. Allen
- Department of Pharmacology, University of Alberta, Edmonton, Canada
- Centre for Drug Research and Development, Vancouver, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, BC Cancer Agency, Vancouver, Canada
- Centre for Drug Research and Development, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Dawn N. Waterhouse
- Experimental Therapeutics, BC Cancer Agency, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
45
|
Uchida K, Otake K, Tanaka K, Hashimoto K, Saigusa S, Matsushita K, Koike Y, Inoue M, Ueeda M, Okugawa Y, Inoue Y, Mohri Y, Kusunoki M. Clinical implications of CES2 RNA expression in neuroblastoma. J Pediatr Surg 2013; 48:502-9. [PMID: 23480903 DOI: 10.1016/j.jpedsurg.2012.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 10/02/2012] [Accepted: 10/03/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND/PURPOSE Human carboxylesterase 2 (CES2) is the key enzyme for metabolic activation of irinotecan (CPT-11). The aim was to evaluate the clinical implications of CES2 RNA expression in neuroblastoma cells. METHODS CES2 RNA expression was determined by real-time reverse transcription-polymerase chain reaction in five neuroblastoma cell lines and 42 clinical samples of untreated neuroblastoma. Sensitivity to CPT-11 was assessed by WST-8 colorimetric assays. Induction of apoptosis was evaluated by flow cytometry after CPT-11 exposure. Protein expression of CES2 was evaluated by Western blotting analysis. CES2 RNA expression in clinical samples was investigated for its associations with the clinicopathological characteristics. RESULTS CES2 RNA expression was observed in neuroblastoma cells, and its expression in neuroblastoma cell lines was positively correlated with sensitivity to CPT-11 and apoptosis after CPT-11 exposure in vitro. CES2 RNA expression was correlated with the protein levels of CES2 in vitro. CES2 RNA expression was significantly higher in patients with a characteristic related to advanced disease. CONCLUSIONS Our results suggest the potential of clinical application of CPT-11 in neuroblastoma treatment for patients with advanced disease.
Collapse
Affiliation(s)
- Keiichi Uchida
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Topoisomerases are nuclear enzymes that play essential roles in DNA replication, transcription, chromosome segregation, and recombination. All cells have two major forms of topoisomerases: type I enzymes, which make single-stranded cuts in DNA, and type II enzymes, which cut and pass double-stranded DNA. DNA topoisomerases are important targets of approved and experimental anti-cancer agents. The protocols described in this unit are for assays used to assess new chemical entities for their ability to inhibit both forms of DNA topoisomerase. Included are an in vitro assay for topoisomerase I activity based on relaxation of supercoiled DNA, and an assay for topoisomerase II based on the decatenation of double-stranded DNA. The preparation of mammalian cell extracts for assaying topoisomerase activity is described, along with a protocol for an ICE assay to examine topoisomerase covalent complexes in vivo, and an assay for measuring DNA cleavage in vitro.
Collapse
Affiliation(s)
- John L Nitiss
- Molecular Pharmacology Department, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | | |
Collapse
|
47
|
Buchler T, Pavlik T, Bortlicek Z, Poprach A, Vyzula R, Abrahamova J, Melichar B. Objective response and time to progression on sequential treatment with sunitinib and sorafenib in metastatic renal cell carcinoma. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2012; 156:81-92. [PMID: 22752573 DOI: 10.5507/bp.2012.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patients with metastatic renal cell carcinoma (mRCC) are often treated sequentially with targeted agents, although the optimal strategy is not known. A retrospective, registry-based study has been carried out to assess correlation between clinical response and progression-free survival in patients with mRCC treated sequentially with tyrosine-kinase inhibitors (TKIs) sunitinib and sorafenib. Data on 218 mRCC patients treated with sunitinib and sorafenib who completed therapy with both TKIs were obtained from a database of mRCC patients. Standard nonparametric methods were used to assess correlation between response, PFS and length of treatment on the two agents. A strong correlation between responses to first- versus second TKI was observed (p < 0.001). No significant association was noted between the duration of therapy with the two TKIs (p = 0.056), although there was a weak statistically significant correlation between progression-free survival times in the subgroup patients who discontinued treatment because of disease progression. In conclusion, the duration of response on first TKI is of limited value in selecting mRCC patients for sequential TKI therapy. There is a strong correlation between the types of tumour response on the first- versus the second TKI.
Collapse
Affiliation(s)
- Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Thomayer Hospital and Charles University, Videnska 800, 140 59, Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Wong CC, Cheng KW, Xie G, Zhou D, Zhu CH, Constantinides PP, Rigas B. Carboxylesterases 1 and 2 hydrolyze phospho-nonsteroidal anti-inflammatory drugs: relevance to their pharmacological activity. J Pharmacol Exp Ther 2012; 340:422-32. [PMID: 22085648 PMCID: PMC3263964 DOI: 10.1124/jpet.111.188508] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 11/04/2011] [Indexed: 12/21/2022] Open
Abstract
Phospho-nonsteroidal anti-inflammatory drugs (phospho-NSAIDs) are novel NSAID derivatives with improved anticancer activity and reduced side effects in preclinical models. Here, we studied the metabolism of phospho-NSAIDs by carboxylesterases and assessed the impact of carboxylesterases on the anticancer activity of phospho-NSAIDs in vitro and in vivo. The expression of human liver carboxylesterase (CES1) and intestinal carboxylesterase (CES2) in human embryonic kidney 293 cells resulted in the rapid intracellular hydrolysis of phospho-NSAIDs. Kinetic analysis revealed that CES1 is more active in the hydrolysis of phospho-sulindac, phospho-ibuprofen, phospho-naproxen, phospho-indomethacin, and phospho-tyrosol-indomethacin that possessed a bulky acyl moiety, whereas the phospho-aspirins are preferentially hydrolyzed by CES2. Carboxylesterase expression leads to a significant attenuation of the in vitro cytotoxicity of phospho-NSAIDs, suggesting that the integrity of the drug is critical for anticancer activity. Benzil and bis-p-nitrophenyl phosphate (BNPP), two carboxylesterase inhibitors, abrogated the effect of carboxylesterases and resensitized carboxylesterase-expressing cells to the potent cytotoxic effects of phospho-NSAIDs. In mice, coadministration of phospho-sulindac and BNPP partially protected the former from esterase-mediated hydrolysis, and this combination more effectively inhibited the growth of AGS human gastric xenografts in nude mice (57%) compared with phospho-sulindac alone (28%) (p = 0.037). Our results show that carboxylesterase mediates that metabolic inactivation of phospho-NSAIDs, and the inhibition of carboxylesterases improves the efficacy of phospho-NSAIDs in vitro and in vivo.
Collapse
Affiliation(s)
- Chi C Wong
- Division of Cancer Prevention, Department of Medicine, Stony Brook University, Stony Brook, New York 11794-8173, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Wang H, Shrestha TB, Basel MT, Dani RK, Seo GM, Balivada S, Pyle MM, Prock H, Koper OB, Thapa PS, Moore D, Li P, Chikan V, Troyer DL, Bossmann SH. Magnetic-Fe/Fe(3)O(4)-nanoparticle-bound SN38 as carboxylesterase-cleavable prodrug for the delivery to tumors within monocytes/macrophages. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2012; 3:444-55. [PMID: 23016149 PMCID: PMC3388369 DOI: 10.3762/bjnano.3.51] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/24/2012] [Indexed: 05/20/2023]
Abstract
The targeted delivery of therapeutics to the tumor site is highly desirable in cancer treatment, because it is capable of minimizing collateral damage. Herein, we report the synthesis of a nanoplatform, which is composed of a 15 ± 1 nm diameter core/shell Fe/Fe(3)O(4) magnetic nanoparticles (MNPs) and the topoisomerase I blocker SN38 bound to the surface of the MNPs via a carboxylesterase cleavable linker. This nanoplatform demonstrated high heating ability (SAR = 522 ± 40 W/g) in an AC-magnetic field. For the purpose of targeted delivery, this nanoplatform was loaded into tumor-homing double-stable RAW264.7 cells (mouse monocyte/macrophage-like cells (Mo/Ma)), which have been engineered to express intracellular carboxylesterase (InCE) upon addition of doxycycline by a Tet-On Advanced system. The nanoplatform was taken up efficiently by these tumor-homing cells. They showed low toxicity even at high nanoplatform concentration. SN38 was released successfully by switching on the Tet-On Advanced system. We have demonstrated that this nanoplatform can be potentially used for thermochemotherapy. We will be able to achieve the following goals: (1) Specifically deliver the SN38 prodrug and magnetic nanoparticles to the cancer site as the payload of tumor-homing double-stable RAW264.7 cells; (2) Release of chemotherapeutic SN38 at the cancer site by means of the self-containing Tet-On Advanced system; (3) Provide localized magnetic hyperthermia to enhance the cancer treatment, both by killing cancer cells through magnetic heating and by activating the immune system.
Collapse
Affiliation(s)
- Hongwang Wang
- Kansas State University, Department of Chemistry, CBC 201, Manhattan, KS 66506
| | - Tej B Shrestha
- Kansas State University, Anatomy & Physiology, Coles 228, Manhattan, KS 66506
| | - Matthew T Basel
- Kansas State University, Anatomy & Physiology, Coles 228, Manhattan, KS 66506
| | - Raj Kumar Dani
- Kansas State University, Department of Chemistry, CBC 201, Manhattan, KS 66506
| | - Gwi-Moon Seo
- Kansas State University, Anatomy & Physiology, Coles 228, Manhattan, KS 66506
| | - Sivasai Balivada
- Kansas State University, Anatomy & Physiology, Coles 228, Manhattan, KS 66506
| | - Marla M Pyle
- Kansas State University, Anatomy & Physiology, Coles 228, Manhattan, KS 66506
| | - Heidy Prock
- Kansas State University, Department of Chemistry, CBC 201, Manhattan, KS 66506
| | - Olga B Koper
- Battelle Memorial Institute, 505 King Ave., Columbus, OH 43201
| | - Prem S Thapa
- University of Kansas, KU Microscopy & Analytical Imaging Laboratory, 1043 Haworth, Lawrence, KS 66045
| | - David Moore
- University of Kansas, KU Microscopy & Analytical Imaging Laboratory, 1043 Haworth, Lawrence, KS 66045
| | - Ping Li
- Kansas State University, Department of Chemistry, CBC 201, Manhattan, KS 66506
| | - Viktor Chikan
- Kansas State University, Department of Chemistry, CBC 201, Manhattan, KS 66506
| | - Deryl L Troyer
- Kansas State University, Anatomy & Physiology, Coles 228, Manhattan, KS 66506
| | - Stefan H Bossmann
- Kansas State University, Department of Chemistry, CBC 201, Manhattan, KS 66506
| |
Collapse
|