1
|
Brennan CK, Yao Z, Ionkina AA, Rathbun CM, Sathishkumar B, Prescher JA. Multiplexed bioluminescence imaging with a substrate unmixing platform. Cell Chem Biol 2022; 29:1649-1660.e4. [PMID: 36283402 PMCID: PMC9675729 DOI: 10.1016/j.chembiol.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/31/2022] [Accepted: 09/30/2022] [Indexed: 01/31/2023]
Abstract
Bioluminescent tools can illuminate cellular features in whole organisms. Multi-component tracking remains challenging, though, owing to a lack of well-resolved probes and long imaging times. To address the need for more rapid, quantitative, and multiplexed bioluminescent readouts, we developed an analysis pipeline featuring sequential substrate administration and serial image acquisition. Light output from each luciferin is layered on top of the previous image, with minimal delay between substrate delivery. A MATLAB algorithm was written to analyze bioluminescent images generated from the rapid imaging protocol and deconvolute (i.e., unmix) signals from luciferase-luciferin pairs. Mixtures comprising three to five luciferase reporters were readily distinguished in under 50 min; this same experiment would require days using conventional workflows. We further showed that the algorithm can be used to accurately quantify luciferase levels in heterogeneous mixtures. Based on its speed and versatility, the multiplexed imaging platform will expand the scope of bioluminescence technology.
Collapse
Affiliation(s)
- Caroline K Brennan
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Zi Yao
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Anastasia A Ionkina
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Colin M Rathbun
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | | | - Jennifer A Prescher
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
2
|
Woradulayapinij W, Pothiluk A, Nualsanit T, Yimsoo T, Yingmema W, Rojanapanthu P, Hong Y, Baek SJ, Treesuppharat W. Acute oral toxicity of damnacanthal and its anticancer activity against colorectal tumorigenesis. Toxicol Rep 2022; 9:1968-1976. [PMID: 36518435 PMCID: PMC9742955 DOI: 10.1016/j.toxrep.2022.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/05/2022] Open
Abstract
Damnacanthal is an anthraquinone, extracted, and purified from the root of Morinda citrifolia in Thailand. This study aimed to measure acute oral toxicity and to investigate the anticancer activity of damnacanthal in colorectal tumorigenesis. We found that the growth of human colorectal cancer cells was inhibited by damnacanthal in a dose- and a time-dependent manner. The growth inhibitory effect of damnacanthal was better than that of 5-FU used as a positive control in colorectal cancer cells, along with the downregulation of cell cycle protein cyclin D1. Similarly, an oral treatment of damnacanthal effectively inhibited the growth of colorectal tumor xenografts in nude mice, which was approximately 2-3-fold higher as compared to 5-FU by tumor size as well as expression of bioluminescence. Furthermore, the study of acute oral toxicity in mice exhibited a relatively low toxicity of damnacanthal with a LD50 cut-off value of 2500 mg/kg according to OECD Guideline 423. These results reveal the potential therapeutic activity of a natural damnacanthal compound as an anti-colorectal cancer drug.
Collapse
Key Words
- 5-FU, 5-Fluorouracil
- ALT, Alanine aminotransferase
- AST, Aspartate aminotransferase
- Acute oral toxicity
- Anticancer activity
- BSA, Bovine serum albumin
- BUN, Blood urea nitrogen
- Colorectal tumorigenesis
- D20, Damnacanthal at 20 mg/kg
- D40, Damnacanthal at 40 mg/kg
- DMSO, Dimethyl sulfoxide
- DPBS, Dulbecco’s phosphate buffered saline
- Damnacanthal
- F20, 5-Fluorouracil at 20 mg/kg
- FBS, Fetal bovine serum
- FTIR, Fourier transform infrared spectroscopy
- IC50, Half-maximal inhibitory concentration
- LD50, Median lethal dose
- MS, Mass spectrometry
- MTT, 3-(4,5-Dimethythiazol-2-yl)− 2,5-diphenyltetrazolium bromide
- NC, Negative control
- NMR, Nuclear magnetic resonance spectroscopy
- PMSF, Phenylmethanesulfonyl fluoride
- TBST, Tris-buffered saline containing 0.05 % Tween 20
- TLC, Thin layer chromatography
- VLC, Vacuum liquid chromatographic method
Collapse
Affiliation(s)
- Warunya Woradulayapinij
- Thammasat University Research Unit in Mechanisms of Drug Action and Molecular Imaging, Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Apipu Pothiluk
- Thammasat University Research Unit in Mechanisms of Drug Action and Molecular Imaging, Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Thararat Nualsanit
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Thunyatorn Yimsoo
- Laboratory Animal Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Werayut Yingmema
- Laboratory Animal Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Pleumchitt Rojanapanthu
- Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| | - Yukyung Hong
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Seung Joon Baek
- Laboratory of Signal Transduction, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Worapapar Treesuppharat
- Thammasat University Research Unit in Mechanisms of Drug Action and Molecular Imaging, Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Pathum Thani 12120, Thailand
| |
Collapse
|
3
|
Pharmacokinetics of Single Domain Antibodies and Conjugated Nanoparticles Using a Hybrid near Infrared Method. Int J Mol Sci 2021; 22:ijms22168695. [PMID: 34445399 PMCID: PMC8395466 DOI: 10.3390/ijms22168695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
Iron oxide nanoparticles and single domain antibodies from camelids (VHHs) have been increasingly recognized for their potential uses for medical diagnosis and treatment. However, there have been relatively few detailed characterizations of their pharmacokinetics (PK). The aim of this study was to develop imaging methods and pharmacokinetic models to aid the future development of a novel family of brain MRI molecular contrast agents. An efficient near-infrared (NIR) imaging method was established to monitor VHH and VHH conjugated nanoparticle kinetics in mice using a hybrid approach: kinetics in blood were assessed by direct sampling, and kinetics in kidney, liver, and brain were assessed by serial in vivo NIR imaging. These studies were performed under "basal" circumstances in which the VHH constructs and VHH-conjugated nanoparticles do not substantially interact with targets nor cross the blood brain barrier. Using this approach, we constructed a five-compartment PK model that fits the data well for single VHHs, engineered VHH trimers, and iron oxide nanoparticles conjugated to VHH trimers. The establishment of the feasibility of these methods lays a foundation for future PK studies of candidate brain MRI molecular contrast agents.
Collapse
|
4
|
Dai Y, Chen D, Wang G, Yin J, Zhan Y, Wu K, Liang J, Chen X. Kinetic modeling and analysis of dynamic bioluminescence imaging of substrates administered by intraperitoneal injection. Quant Imaging Med Surg 2020; 10:389-396. [PMID: 32190565 DOI: 10.21037/qims.2020.01.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Bioluminescence imaging (BLI) has been found to have diverse applications in the life sciences and medical research due to its ease of use and high sensitivity. From kinetics analysis, dynamic imaging studies have significant advantages for diagnosis when compared to traditional static imaging studies. This work focuses on modeling and quantitatively analyzing the dynamic data produced from the intraperitoneal (IP) injection of D-luciferin in longitudinal BLI, aiming to provide a powerful tool for monitoring the growth of tumors. Methods We constructed a three-compartment pharmacokinetic (PK) model and employed the standard Michaelis-Menten (M-M) kinetics to investigate the dynamic BLI data produced from the IP injection of D-luciferin. The 3 compartments were the plasma compartment, the non-specific compartment, and the specific compartment. The validity of this PK model was tested by the dynamic BLI data of MKN28M-luc xenograft mice, along with the published longitudinal dynamic BLI data of B16F10-luc xenograft mice. Results The R-squares between the simulated lines and the measurement were 1 and 0.99, respectively, for the mice data and the published data. In addition, the 2 kinetic macroparameters obtained reflected the rate of tumor growth in vivo. In particular, the values of macroparameters A showed a significant dependence on tumor surface area. Conclusions The proposed PK model may be an effective tool for use in drug development programs and for monitoring the response of tumors to treatment.
Collapse
Affiliation(s)
- Yunpeng Dai
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an 710071, China
| | - Duofang Chen
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an 710071, China
| | - Guodong Wang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jipeng Yin
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yonghua Zhan
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an 710071, China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jimin Liang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an 710071, China
| | - Xueli Chen
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education & School of Life Science and Technology, Xidian University, Xi'an 710071, China
| |
Collapse
|
5
|
Coix Seed Extract Enhances the Anti-Pancreatic Cancer Efficacy of Gemcitabine through Regulating ABCB1- and ABCG2-Mediated Drug Efflux: A Bioluminescent Pharmacokinetic and Pharmacodynamic Study. Int J Mol Sci 2019; 20:ijms20215250. [PMID: 31652737 PMCID: PMC6862065 DOI: 10.3390/ijms20215250] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 01/29/2023] Open
Abstract
A deep insight into the function and kinetics of ATP-binding cassette (ABC) transporters may aid in the development of pharmaceutics that can minimize the particular facet of chemo-resistance. We utilized bioluminescence imaging to monitor the ABC transporter mediated intracellular drug efflux function. We also investigated the potential association between the intracellular bioluminescent pharmacokinetic profiles and the anti-tumor efficacy of the coix seed extract and gemcitabine against pancreatic cancer cells in vitro and in vivo. The bioluminescent pharmacokinetic parameters and pharmacodynamic index (IC50 and TGI) were determined. The expression levels ABCB1 and ABCG2 were assessed. Results showed that coix seed extract could synergistically enhance the anti-cancer efficacy of gemcitabine (p < 0.05). Meanwhile coix seed extract alone or in combination with gemcitabine could significantly increase the AUCluc while decreasing the Kluc (p < 0.01). Western blot and immunohistochemistry assay demonstrated that coix seed extract could significantly mitigate gemcitabine-induced upregulation of ABCB1 and ABCG2 protein. The Pearson correlation analysis demonstrated that the bioluminescent pharmacokinetic parameters and pharmacodynamic index have strong association in vitro and in vivo. In conclusion coix seed extract could augment the efficacy of gemcitabine therapy in pancreatic cancer cells may at least partly due to the alteration of ABC transporter-mediated drug efflux function.
Collapse
|
6
|
Pharmacodynamic modelling of resistance to epidermal growth factor receptor inhibition in brain metastasis mouse models. Cancer Chemother Pharmacol 2018; 82:669-675. [PMID: 30054711 PMCID: PMC6132866 DOI: 10.1007/s00280-018-3630-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023]
Abstract
Purpose Epidermal growth factor receptor (EGFR) is thought to play a role in the regulation of cell proliferation; with its activation stimulating tumour growth. EGFR inhibitors have shown promise in the treatment of cancer, particularly in non-small cell lung cancer, however, resistance is observed in the majority of patients. A tumour growth model was developed aiming to explain this resistance. Methods The model incorporating populations of both sensitive and resistant cells were fitted to data from a study of EGFR inhibitor AZD3759 in brain metastasis mouse models. The observed regrowth of tumours in higher dose groups suggested the development of resistance to treatment. The bioluminescence observations were highly variable, covering many orders of magnitude, so to assess how reliable the model was, the parameter estimates were compared to those found in less noisy subcutaneous mouse models. Results The fitted model suggested that resistance was mainly due to a proportion of cells being resistant at baseline, and the contribution of mutations occurring during the study leading to resistance was negligible. Estimated growth rate and dose–response was found to be comparable between brain metastasis and subcutaneous mouse models. Conclusions The developed model to describe resistance suggests that the resistance to EGFR-inhibition seen in these xenografts is best described by assuming a small percentage of cells are resistant to treatment at baseline. This model suggests changes to dosing and dosing schedule may not prevent resistance to treatment developing, and that additional treatments would need to be used in combination to overcome resistance.
Collapse
|
7
|
Dai Y, Wang G, Chen D, Yin J, Zhan Y, Nie Y, Wu K, Liang J, Chen X. Intravenous Administration-Oriented Pharmacokinetic Model for Dynamic Bioluminescence Imaging. IEEE Trans Biomed Eng 2018; 66:843-847. [PMID: 30047868 DOI: 10.1109/tbme.2018.2858774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE In vivo bioluminescence imaging (BLI) is a promising tool for monitoring the growth and metastasis of tumors. However, quantitative BLI research based on intravenous (IV) injection is limited, which greatly restricts its further application. To address this problem, we designed a pharmacokinetic (PK) model which is suitable for applying on IV administration of small amounts of D-Luciferin. METHODS After three weeks of postimplantation, mkn28-luc xenografted mice were subjected to 40-min dynamic BLI immediately following D-Luciferin intravenous injection on days 1, 3, 5, 7, and 9. Furthermore, the PK model was applied on dynamic BLI data to obtain the sum of kinetic rate constants (SKRC). RESULTS Results showed that the SKRC values decreased rapidly with the growth of the tumor. There was a statistical difference between the SKRC values measured at different time points, while the time point of luminous intensity peak was unaffected by the growth of the tumor. CONCLUSION In short, our results imply that dynamic BLI combined with our PK model can predict tumor growth earlier and with higher sensitivity compared to the conventional method, which is crucial for improving drug evaluation efficacy. In addition, the dynamic BLI may provide a valuable reference for the noninvasive acquiring arterial input function, which may also provide a new application prospect for hybrid PET-optical imaging.
Collapse
|
8
|
Tang CY, Zhu LX, Yu JD, Chen Z, Gu MC, Mu CF, Liu Q, Xiong Y. Effect of β-elemene on the kinetics of intracellular transport of d-luciferin potassium salt (ABC substrate) in doxorubicin-resistant breast cancer cells and the associated molecular mechanism. Eur J Pharm Sci 2018; 120:20-29. [PMID: 29704644 DOI: 10.1016/j.ejps.2018.04.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/06/2018] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
In order to explore the mechanism of the reversing multidrug resistance (MDR) phenotypes by β-elemene (β-ELE) in doxorubicin (DOX)-resistant breast cancer cells (MCF-7/DOX), both the functionality and quantity of the ABC transporters in MCF-7/DOX were studied. Bioluminescence imaging (BLI) was used to study the efflux of d-luciferin potassium salt, the substrate of ATP-binding cassette transporters (ABC transporters), in MCF-7/DOX cells treated by β-ELE. At the same time three major ABC transport proteins and genes-related MDR, P-glycoprotein (P-gp, ABCB1) and multidrug resistance-associated protein 1 (MRP, ABCC1) as well as breast cancer resistance protein (BCRP, ABCG2) were analyzed by q-PCR and Western blot. To investigate the efflux functionality of ABC transporters, MCF-7/DOXFluc cell line with stably-overexpressed luciferase was established. BLI was then used to real-time monitor the efflux kinetics of d-luciferin potassium salt before and after MCF-7/DOXFluc cells being treated with β-ELE or not. The results showed that the efflux of d-luciferin potassium salt from MCF-7/DOXFluc was lessened when pretreated with β-ELE, which means that β-ELE may dampen the functionality of ABC transporters, thus decrease the efflux of d-fluorescein potassium or other chemotherapies which also serve as the substrates of ABC transporters. As the effect of β-ELE on the expression of ABC transporters, the results of q-PCR and Western blot showed that gene and protein expression of ABC transporters such as P-gp, MRP, and BCRP were down-regulated after the treatment of β-ELE. To verify the efficacy of β-ELE on reversing MDR, MCF-7/DOX cells were treated with the combination of DOX and β-ELE. MTT assay showed that β-ELE increased the inhibitory effect of DOX on the proliferation of MCF-7/DOX, and the IC50 of the combination group was much lower than that of the single DOX or β-ELE treatment. In all, β-ELE may reverse MDR through the substrates of ABC transporters by two ways, to lessen the ABC protein efflux by weakening their functionality, or to reduce the quantity of ABC gene and protein expression.
Collapse
Affiliation(s)
- Chao-Yuan Tang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Li-Xin Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Zhejiang Institute for Food and Drug Control, Hangzhou, Zhejiang 310004, China
| | - Jian-Dong Yu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Zhi Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Man-Cang Gu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Chao-Feng Mu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill 27599, NC, USA
| | - Yang Xiong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
9
|
Buchakjian MR, Merritt NM, Moose DL, Dupuy AJ, Tanas MR, Henry MD. A Trp53fl/flPtenfl/fl mouse model of undifferentiated pleomorphic sarcoma mediated by adeno-Cre injection and in vivo bioluminescence imaging. PLoS One 2017; 12:e0183469. [PMID: 28841687 PMCID: PMC5571905 DOI: 10.1371/journal.pone.0183469] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 08/04/2017] [Indexed: 12/24/2022] Open
Abstract
Genetic mouse models of soft tissue sarcoma provide critical insights into disease pathophysiology, which are oftentimes unable to be extracted from human tumor samples or xenograft models. In this study we describe a mouse model of soft tissue sarcoma mediated by adenoviral-Cre recombinase injection into Trp53fl/fl/Ptenfl/fl lox-stop-lox luciferase mice. Injection of adenovirus expressing Cre recombinase, either subcutaneously or intramuscularly in two experimental groups, results in viral infection and gene recombination with 100% penetrance within the first 24 hours following injection. Luciferase expression measured by real-time bioluminescence imaging increases over time, with an initial robust increase following viral injection, followed by a steady rise over the next several weeks as primary tumors develop and grow. Intramuscular injections were more commonly associated with evidence of systemic viral distribution than subcutaneous injections. All mice developed soft tissue sarcomas at the primary injection site, with histological examination identifying 93% of tumors as invasive pleomorphic sarcomas based on microscopic morphology and immunohistochemical expression of sarcoma markers. A lymphocytic infiltrate was present in 64% of the sarcomas in this immunocompetent model and 71% of tumors expressed PD-L1. This is the first report of a viral-Cre mediated Trp53/Pten mouse model of undifferentiated pleomorphic sarcoma. The bioluminescence imaging feature, along with high penetrance of the model and its immunological characteristics, makes it suited for pre-clinical studies of soft tissue sarcoma.
Collapse
Affiliation(s)
- Marisa R. Buchakjian
- Department of Otolaryngology – Head and Neck Surgery, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
| | - Nicole M. Merritt
- Department of Pathology, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
| | - Devon L. Moose
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Adam J. Dupuy
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Munir R. Tanas
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- * E-mail:
| | - Michael D. Henry
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
| |
Collapse
|
10
|
Gemcitabine upregulates ABCG2/BCRP and modulates the intracellular pharmacokinetic profiles of bioluminescence in pancreatic cancer cells. Anticancer Drugs 2016; 27:183-91. [PMID: 26556627 DOI: 10.1097/cad.0000000000000315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A lack of methods capable of exploring real-time intracellular drug deposition has since limited the investigation of gemcitabine-induced multidrug resistance in vitro and in vivo. Specifically, resistance induced by D-luciferin, a substrate of the breast cancer resistance protein (ABCG2/BCRP), has recently attracted clinical attention, but further investigation has been limited. Herein, the intracellular pharmacokinetic behavior of D-luciferin was investigated in pancreatic cancer cell lines in real time by using bioluminescence imaging. To achieve this feat, BxPC3 and Panc1 pancreatic cancer cells overexpressing firefly luciferase were treated with gemcitabine in a dose and time gradient manner in vitro. The intracellular pharmacokinetic profiles of each group were then determined through the acquisition of bioluminescent signal intensity of D-luciferin in cells. Simultaneously, key pharmacokinetic parameters including area under the curve, elimination rate constant (K), and mean resident time were calculated according to the noncompartment model. ABCG2 protein levels following gemcitabine treatment were detected through western blot, and gemcitabine showed no significant effect on luciferase activity over dimethyl sulfoxide (DMSO) as a control (P>0.05). However, gemcitabine significantly increased K values while suppressing area under the curve and mean resident time compared with DMSO (P<0.05) and increased ABCG2 expression over DMSO-treated cells. In addition, gemcitabine increased the elimination rate of the ABCG2 substrate, D-luciferin, and decreased D-luciferin accumulation in BxPC3 and Panc1 cells in a dose-response manner. Advances made herein illustrate the versatility of the in-vitro bioluminescent model and its capability to observe the onset of chemoresistance in real time.
Collapse
|
11
|
Christensen J, Vonwil D, Shastri VP. Non-Invasive In Vivo Imaging and Quantification of Tumor Growth and Metastasis in Rats Using Cells Expressing Far-Red Fluorescence Protein. PLoS One 2015; 10:e0132725. [PMID: 26186005 PMCID: PMC4505884 DOI: 10.1371/journal.pone.0132725] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/17/2015] [Indexed: 01/26/2023] Open
Abstract
Non-invasive in vivo imaging is emerging as an important tool for basic and preclinical research. Near-infrared (NIR) fluorescence dyes and probes have been used for non-invasive optical imaging since in the NIR region absorption and auto fluorescence by body tissue is low, thus permitting for greater penetration depths and high signal to noise ratio. Currently, cell tracking systems rely on labeling cells prior to injection or administering probes targeting the cell population of choice right before imaging. These approaches do not enable imaging of tumor growth, as the cell label is diluted during cell division. In this study we have developed cell lines stably expressing the far-red fluorescence protein E2-Crimson, thus enabling continuous detection and quantification of tumor growth. In a xenograft rat model, we show that E2-Crimson expressing cells can be detected over a 5 week period using optical imaging. Fluorescence intensities correlated with tumor volume and weight and allowed for a reliable and robust quantification of the entire tumor compartment. Using a novel injection regime, the seeding of MDA-MB-231 breast cancer cells in the lungs in a rat model was established and verified.
Collapse
Affiliation(s)
- Jon Christensen
- Institute for Macromolecular Chemistry, University of Freiburg, 79104, Freiburg, Germany
- BIOSS—Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Daniel Vonwil
- Institute for Macromolecular Chemistry, University of Freiburg, 79104, Freiburg, Germany
| | - V. Prasad Shastri
- Institute for Macromolecular Chemistry, University of Freiburg, 79104, Freiburg, Germany
- BIOSS—Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| |
Collapse
|
12
|
Abstract
Pancreatic cancer is an insidious type of cancer with its symptoms manifested upon extensive disease. The overall 5-year survival rates between 0.4 and 4%. Surgical resection is an option for only 10% of the patients with pancreatic cancer. Local recurrence and hepatic metastases occur within 2 years after surgery. There are currently several molecular pathways investigated and novel targeted treatments are on the market. However; the nature of pancreatic cancer with its ability to spread locally in the primary site and lymph nodes indicates that further experimentation with local interventional therapies could be a future treatment proposal as palliative care or adjunct to gene therapy and chemotherapy/radiotherapy. In the current review, we will summarize the molecular pathways and present the interventional treatment options for pancreatic cancer.
Collapse
|
13
|
Hohenforst-Schmidt W, Zarogoulidis P, Stopek J, Vogl T, Hübner F, Turner JF, Browning R, Zarogoulidis K, Drevelegas A, Drevelegas K, Darwiche K, Freitag L, Rittger H. DDMC-p53 gene therapy with or without cisplatin and microwave ablation. Onco Targets Ther 2015; 8:1165-73. [PMID: 26056480 PMCID: PMC4446017 DOI: 10.2147/ott.s83794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lung cancer remains the leading cause of death in cancer patients. Severe treatment side effects and late stage of disease at diagnosis continue to be an issue. We investigated whether local treatment using 2-diethylaminoethyl-dextran methyl methacrylate copolymer with p53 (DDMC-p53) with or without cisplatin and/or microwave ablation enhances disease control in BALBC mice. We used a Lewis lung carcinoma cell line to inoculate 140 BALBC mice, which were divided into the following seven groups; control, cisplatin, microwave ablation, DDMC-p53, DDMC-p53 plus cisplatin, DDMC-p53 plus microwave, and DDMC-p53 plus cisplatin plus microwave. Microwave ablation energy was administered at 20 W for 10 minutes. Cisplatin was administered as 1 mL/mg and the DDMC-p53 complex delivered was 0.5 mL. Increased toxicity was observed in the group receiving DDMC-p53 plus cisplatin plus microwave followed by the group receiving DDMC-p53 plus cisplatin. Infection after repeated treatment administration was a major issue. We conclude that a combination of gene therapy using DDMC-p53 with or without cisplatin and microwave is an alternative method for local disease control. However, more experiments are required in a larger model to identify the appropriate dosage profile.
Collapse
Affiliation(s)
| | - Paul Zarogoulidis
- Pulmonary Department-Oncology Unit, G Papanikolaou General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Thomas Vogl
- Department of Diagnostic and Interventional Radiology, Goethe University of Frankfurt, Frankfurt, Germany
| | - Frank Hübner
- II Medical Clinic, Coburg Hospital, University of Wuerzburg, Coburg, Germany
| | - J Francis Turner
- Division of Interventional Pulmonology, Western Regional Medical Center, Goodyear, AZ ; Medical Oncology, Cancer Treatment Centers of America, Western Regional Medical Center, Goodyear, AZ
| | - Robert Browning
- Pulmonary and Critical Care Medicine, Interventional Pulmonology, National Naval Medical Center, Walter Reed Army Medical Center, Bethesda, MD, USA
| | - Konstantinos Zarogoulidis
- Pulmonary Department-Oncology Unit, G Papanikolaou General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonis Drevelegas
- Radiology Department, Interbalkan European Medical Center, Thessaloniki, Greece
| | | | - Kaid Darwiche
- Department of interventional Pneumology, Ruhrlandklinik, University Hospital Essen, University of Essen-Duisburg, Essen, Germany
| | - Lutz Freitag
- Department of interventional Pneumology, Ruhrlandklinik, University Hospital Essen, University of Essen-Duisburg, Essen, Germany
| | - Harald Rittger
- Medical Clinic I, 'Fuerth Hospital, University of Erlangen, Erlangen, Germany
| |
Collapse
|
14
|
Wang L, An Y, Yuan C, Zhang H, Liang C, Ding F, Gao Q, Zhang D. GEM-loaded magnetic albumin nanospheres modified with cetuximab for simultaneous targeting, magnetic resonance imaging, and double-targeted thermochemotherapy of pancreatic cancer cells. Int J Nanomedicine 2015; 10:2507-19. [PMID: 25848268 PMCID: PMC4386779 DOI: 10.2147/ijn.s77642] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Targeted delivery is a promising strategy to improve the diagnostic imaging and therapeutic effect of cancers. In this paper, novel cetuximab (C225)-conjugated, gemcitabine (GEM)-containing magnetic albumin nanospheres (C225-GEM/MANs) were fabricated and applied as a theranostic nanocarrier to conduct simultaneous targeting, magnetic resonance imaging (MRI), and double-targeted thermochemotherapy against pancreatic cancer cells. Methods Fe3O4 nanoparticles (NPs) and GEM co-loaded albumin nanospheres (GEM/MANs) were prepared, and then C225 was further conjugated to synthesize C225-GEM/MANs. Their morphology, mean particle size, GEM encapsulation ratio, specific cell-binding ability, and thermal dynamic profiles were characterized. The effects of discriminating different EGFR-expressing pancreatic cancer cells (AsPC-1 and MIA PaCa-2) and monitoring cellular targeting effects were assessed by targeted MRI. Lastly, the antitumor efficiency of double/C225/magnetic-targeted and nontargeted thermochemotherapy was compared with chemotherapy alone using 3-(4, 5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) and flow cytometry (FCM) assay. Results When treated with targeted nanospheres, AsPC-1 cells showed a significantly less intense MRI T2 signal than MIA PaCa-2 cells, while both cells had similar signal strength when incubated with nontargeted nanospheres. T2 signal intensity was significantly lower when magnetic and C225 targeting were combined, rather than used alone. The inhibitory and apoptotic rates of each thermochemotherapy group were significantly higher than those of the chemotherapy-alone groups. Additionally, both MTT and FCM analysis verified that double-targeted thermochemotherapy had the highest targeted killing efficiency among all groups. Conclusion The C225-GEM/MANs can distinguish various EGFR-expressing live pancreatic cancer cells, monitor diverse cellular targeting effects using targeted MRI imaging, and efficiently mediate double-targeted thermochemotherapy against pancreatic cancer cells.
Collapse
Affiliation(s)
- Ling Wang
- Department of Ultrasonography, Zhong Da Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Yanli An
- Medical School, Southeast University, Nanjing, People's Republic of China
| | - Chenyan Yuan
- Department of Clinical Laboratory, Zhong Da Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Hao Zhang
- Medical School, Southeast University, Nanjing, People's Republic of China
| | - Chen Liang
- Medical School, Southeast University, Nanjing, People's Republic of China
| | - Fengan Ding
- Medical School, Southeast University, Nanjing, People's Republic of China
| | - Qi Gao
- Department of Ultrasonography, Zhong Da Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Dongsheng Zhang
- Jiangsu Key Laboratory for Biomaterials and Devices, Medical School, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
15
|
Lambrechts D, Roeffaers M, Kerckhofs G, Hofkens J, Van de Putte T, Schrooten J, Van Oosterwyck H. Reporter cell activity within hydrogel constructs quantified from oxygen-independent bioluminescence. Biomaterials 2014; 35:8065-77. [PMID: 24957291 DOI: 10.1016/j.biomaterials.2014.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/01/2014] [Indexed: 12/12/2022]
Abstract
By providing a three-dimensional (3D) support to cells, hydrogels offer a more relevant in vivo tissue-like environment as compared to two-dimensional cell cultures. Hydrogels can be applied as screening platforms to investigate in 3D the role of biochemical and biophysical cues on cell behaviour using bioluminescent reporter cells. Gradients in oxygen concentration that result from the interplay between molecular transport and cell metabolism can however cause substantial variability in the observed bioluminescent reporter cell activity. To assess the influence of these oxygen gradients on the emitted bioluminescence for various hydrogel geometries, a combined experimental and modelling approach was implemented. We show that the applied model is able to predict oxygen gradient independent bioluminescent intensities which correlate better to the experimentally determined viable cell numbers, as compared to the experimentally measured bioluminescent intensities. By analysis of the bioluminescence reaction dynamics we obtained a quantitative description of cellular oxygen metabolism within the hydrogel, which was validated by direct measurements of oxygen concentration within the hydrogel. Bioluminescence peak intensities can therefore be used as a quantitative measurement of reporter cell activity within a hydrogel, but an unambiguous interpretation of these intensities requires a compensation for the influence of cell-induced oxygen gradients on the luciferase activity.
Collapse
Affiliation(s)
- Dennis Lambrechts
- Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44 - Box 2450, 3001 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering Leuven, KU Leuven, Herestraat 49 - Box 813, 3000 Leuven, Belgium
| | - Maarten Roeffaers
- Center for Surface Chemistry and Catalysis, KU Leuven, Kasteelpark Arenberg 23, 3001 Leuven, Belgium
| | - Greet Kerckhofs
- Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44 - Box 2450, 3001 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering Leuven, KU Leuven, Herestraat 49 - Box 813, 3000 Leuven, Belgium
| | - Johan Hofkens
- Molecular Imaging and Photonics, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Tom Van de Putte
- TiGenix NV, Haasrode Researchpark 1724, Romeinse Straat 12 box 2, 3001 Leuven, Belgium
| | - Jan Schrooten
- Department of Materials Engineering, KU Leuven, Kasteelpark Arenberg 44 - Box 2450, 3001 Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering Leuven, KU Leuven, Herestraat 49 - Box 813, 3000 Leuven, Belgium.
| | - Hans Van Oosterwyck
- Prometheus, Division of Skeletal Tissue Engineering Leuven, KU Leuven, Herestraat 49 - Box 813, 3000 Leuven, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C - Box 2419, 3001 Leuven, Belgium.
| |
Collapse
|
16
|
Johnson CH, Fisher TS, Hoang LT, Felding BH, Siuzdak G, O’Brien PJ. Luciferase does not Alter Metabolism in Cancer Cells. Metabolomics 2014; 10:354-360. [PMID: 24791164 PMCID: PMC4002053 DOI: 10.1007/s11306-014-0622-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Luciferase transfected cell lines are used extensively for cancer models, revealing valuable biological information about disease mechanisms. However, these genetically encoded reporters, while useful for monitoring tumor response in cancer models, can impact cell metabolism. Indeed firefly luciferase and fatty acyl-CoA synthetases differ by a single amino acid, raising the possibility that luciferase activity might alter metabolism and introduce experimental artifacts. Therefore knowledge of the metabolic response to luciferase transfection is of significant importance, especially given the thousands of research studies using luciferase as an in vivo bioluminescence imaging (BLI) reporter. Untargeted metabolomics experiments were performed to examine three different types of lymphoblastic leukemia cell lines (Ramos, Raji and SUP T1) commonly used in cancer research, each were analyzed with and without vector transduction. The Raji model was also tested under perturbed starvation conditions to examine potential luciferase-mediated stress responses. The results showed that no significant metabolic differences were observed between parental and luciferase transduced cells for each cell line, and that luciferase overexpression does not alter cell metabolism under basal or perturbed conditions.
Collapse
Affiliation(s)
- Caroline H. Johnson
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, La Jolla, CA, USA
| | - Timothy S. Fisher
- Pfizer Worldwide Research and Development, La Jolla Laboratories 10724 Science Center Drive, San Diego, CA, USA
| | - Linh T. Hoang
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, La Jolla, CA, USA
| | - Brunhilde H. Felding
- Departments of Molecular and Experimental Medicine and Chemical Physiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics and Mass Spectrometry, The Scripps Research Institute, La Jolla, CA, USA
- Correspondence to: and
| | - Peter J. O’Brien
- Pfizer Worldwide Research and Development, La Jolla Laboratories 10724 Science Center Drive, San Diego, CA, USA
- Correspondence to: and
| |
Collapse
|
17
|
Intratumoral gene therapy versus intravenous gene therapy for distant metastasis control with 2-diethylaminoethyl-dextran methyl methacrylate copolymer non-viral vector-p53. Gene Ther 2013; 21:158-67. [PMID: 24285215 DOI: 10.1038/gt.2013.68] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/06/2013] [Accepted: 10/17/2013] [Indexed: 12/18/2022]
Abstract
Lung cancer still remains to be challenged by novel treatment modalities. Novel locally targeted routes of administration are a methodology to enhance treatment and reduce side effects. Intratumoral gene therapy is a method for local treatment and could be used either in early-stage lung cancer before surgery or at advanced stages as palliative care. Novel non-viral vectors are also in demand for efficient gene transfection to target local cancer tissue and at the same time protect the normal tissue. In the current study, C57BL/6 mice were divided into three groups: (a) control, (b) intravenous and (c) intatumoral gene therapy. The novel 2-Diethylaminoethyl-Dextran Methyl Methacrylate Copolymer Non-Viral Vector (Ryujyu Science Corporation) was conjugated with plasmid pSicop53 from the company Addgene for the first time. The aim of the study was to evaluate the safety and efficacy of targeted gene therapy in a Lewis lung cancer model. Indeed, although the pharmacokinetics of the different administration modalities differs, the intratumoral administration presented increased survival and decreased distant metastasis. Intratumoral gene therapy could be considered as an efficient local therapy for lung cancer.
Collapse
|
18
|
Hohenforst-Schmidt W, Zarogoulidis P, Darwiche K, Vogl T, Goldberg EP, Huang H, Simoff M, Li Q, Browning R, Turner FJ, Le Pivert P, Spyratos D, Zarogoulidis K, Celikoglu SI, Celikoglu F, Brachmann J. Intratumoral chemotherapy for lung cancer: re-challenge current targeted therapies. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:571-83. [PMID: 23898222 PMCID: PMC3718837 DOI: 10.2147/dddt.s46393] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Strategies to enhance the already established doublet chemotherapy regimen for lung cancer have been investigated for more than 20 years. Initially, the concept was to administer chemotherapy drugs locally to the tumor site for efficient diffusion through passive transport within the tumor. Recent advances have enhanced the diffusion of pharmaceuticals through active transport by using pharmaceuticals designed to target the genome of tumors. In the present study, five patients with non-small cell lung cancer epidermal growth factor receptor (EGFR) negative stage IIIa–IV International Union Against Cancer 7 (UICC-7), and with Eastern Cooperative Oncology Group (ECOG) 2 scores were administered platinum-based doublet chemotherapy using combined intratumoral-regional and intravenous route of administration. Cisplatin analogues were injected at 0.5%–1% concentration within the tumor lesion and proven malignant lymph nodes according to pretreatment histological/cytological results and the concentration of systemic infusion was decreased to 70% of a standard protocol. This combined intravenous plus intratumoral-regional chemotherapy is used as a first line therapy on this short series of patients. To the best of our knowledge this is the first report of direct treatment of involved lymph nodes with cisplatin by endobronchial ultrasound drug delivery with a needle without any adverse effects. The initial overall survival and local response are suggestive of a better efficacy compared to established doublet cisplatin–based systemic chemotherapy in (higher) standard concentrations alone according to the UICC 7 database expected survival. An extensive search of the literature was performed to gather information of previously published literature of intratumoral chemo-drug administration and formulation for this treatment modality. Our study shows a favorable local response, more than a 50% reduction, for a massive tumor mass after administration of five sessions of intratumoral chemotherapy plus two cycles of low-dose intravenous chemotherapy according to our protocol. These encouraging results (even in very sick ECOG 2 patients with central obstructive non-small cell lung cancer having a worse prognosis and quality of life than a non-small cell lung cancer in ECOG 0 of the same tumor node metastasis [TNM]-stage without central obstruction) for a chemotherapy-only protocol that differs from conventional cisplatin-based doublet chemotherapy by the route, target site, and dose paves the way for broader applications of this technique. Finally, future perspectives of this treatment and pharmaceutical design for intratumoral administration are presented.
Collapse
|
19
|
Fetterly GJ, Aras U, Lal D, Murphy M, Meholick PD, Wang ES. Development of a preclinical PK/PD model to assess antitumor response of a sequential aflibercept and doxorubicin-dosing strategy in acute myeloid leukemia. AAPS JOURNAL 2013; 15:662-73. [PMID: 23550025 DOI: 10.1208/s12248-013-9480-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 03/21/2013] [Indexed: 12/27/2022]
Abstract
Timing of the anti-angiogenic agent with respect to the chemotherapeutic agent may be crucial in determining the success of combination therapy in cancer. We investigated the effects of sequential therapy with the potent VEGF inhibitor, aflibercept, and doxorubicin (DOX) in preclinical acute myeloid leukemia (AML) models. Mice were engrafted with human HL-60 and HEL-luciferase leukemia cells via S.C. and/or I.V. injection and treated with two to three doses of aflibercept (5-25 mg/kg) up to 3-7 days prior to doxorubicin (30 mg/kg) administration. Leukemia growth was determined by local tumor measurements (days 0-16) and systemic bioluminescent imaging (days 0-28) in animals receiving DOX (3 mg/kg) with or without aflibercept. A PK/PD model was developed to characterize how prior administration of aflibercept altered intratumoral DOX uptake. DOX concentration-time profiles were described using a four-compartment PK model with linear elimination. We determined that intratumoral DOX concentrations were 6-fold higher in the aflibercept plus DOX treatment group versus DOX alone in association with increased drug uptake rates (from 0.125 to 0.471 ml/h/kg) into tumor without affecting drug efflux. PD modeling demonstrated that the observed growth retardation was mainly due to the combination of DOX plus TRAP group; 0.00794 vs. 0.0043 h(-1). This PK/PD modeling approach in leukemia enabled us to predict the effects of dosing frequency and sequence for the combination of anti-VEGF and cytotoxic agents on AML growth in both xenograft and marrow, and may be useful in the design of future rational combinatorial dosing regimens in hematological malignancies.
Collapse
Affiliation(s)
- Gerald J Fetterly
- PK/PD Core Facility, Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Zhang Y, Pullambhatla M, Laterra J, Pomper MG. Influence of bioluminescence imaging dynamics by D-luciferin uptake and efflux mechanisms. Mol Imaging 2012; 11:499-506. [PMID: 23084250 PMCID: PMC4332814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
Abstract
Bioluminescence imaging (BLI) detects light generated by luciferase-mediated oxidation of substrate and is used widely for evaluating transgene expression in cell-based assays and in vivo in relevant preclinical models. The most commonly used luciferase for in vivo applications is firefly luciferase (fLuc), for which D-luciferin serves as the substrate. We demonstrated previously that the expression of the ABCG2 efflux transporter can significantly reduce BLI signal output and that HhAntag-691 can inhibit the efflux of D-luciferin, thereby enhancing BLI signal. Here we show that an HhAntag-691-sensitive uptake mechanism facilitates the intracellular concentration of D-luciferin and that the BLI dynamics of different cell lines are coregulated by this uptake mechanism in conjunction with ABCG2-mediated efflux. After administration of D-luciferin, the HhAntag-691-sensitive uptake mechanism generates a rapid increase in BLI signal that decreases over time, whereas ABCG2-mediated efflux stably reduces signal output. We implicate SLC22A4 (OCTN1), a member of the organic cation/zwitterion uptake transporter family, as one potential mediator of the HhAntag-691-sensitive D-luciferin uptake. These findings provide insight into mechanisms that contribute to the cellular uptake kinetics and in vivo biodistribution of D-luciferin.
Collapse
Affiliation(s)
- Yimao Zhang
- Russell H. Morgan Department of Radiology and Department of Neurology, Johns Hopkins Medical School, Baltimore, MD, USA
| | | | | | | |
Collapse
|
21
|
Zhang Y, Pullambhatla M, Laterra J, Pomper MG. Influence of Bioluminescence Imaging Dynamics by D-Luciferin Uptake and Efflux Mechanisms. Mol Imaging 2012. [DOI: 10.2310/7290.2012.00005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Yimao Zhang
- From the Russell H. Morgan Department of Radiology and Department of Neurology, Johns Hopkins Medical School, and the Kennedy Krieger Institute, Baltimore, MD
| | - Mrudula Pullambhatla
- From the Russell H. Morgan Department of Radiology and Department of Neurology, Johns Hopkins Medical School, and the Kennedy Krieger Institute, Baltimore, MD
| | - John Laterra
- From the Russell H. Morgan Department of Radiology and Department of Neurology, Johns Hopkins Medical School, and the Kennedy Krieger Institute, Baltimore, MD
| | - Martin G. Pomper
- From the Russell H. Morgan Department of Radiology and Department of Neurology, Johns Hopkins Medical School, and the Kennedy Krieger Institute, Baltimore, MD
| |
Collapse
|
22
|
Lin X, Gao R, Zhang Y, Qi N, Zhang Y, Zhang K, He H, Tang X. Lipid nanoparticles for chemotherapeutic applications: strategies to improve anticancer efficacy. Expert Opin Drug Deliv 2012; 9:767-81. [DOI: 10.1517/17425247.2012.685933] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
23
|
Fan TWM, Lorkiewicz PK, Sellers K, Moseley HNB, Higashi RM, Lane AN. Stable isotope-resolved metabolomics and applications for drug development. Pharmacol Ther 2012; 133:366-91. [PMID: 22212615 PMCID: PMC3471671 DOI: 10.1016/j.pharmthera.2011.12.007] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 12/14/2022]
Abstract
Advances in analytical methodologies, principally nuclear magnetic resonance spectroscopy (NMR) and mass spectrometry (MS), during the last decade have made large-scale analysis of the human metabolome a reality. This is leading to the reawakening of the importance of metabolism in human diseases, particularly cancer. The metabolome is the functional readout of the genome, functional genome, and proteome; it is also an integral partner in molecular regulations for homeostasis. The interrogation of the metabolome, or metabolomics, is now being applied to numerous diseases, largely by metabolite profiling for biomarker discovery, but also in pharmacology and therapeutics. Recent advances in stable isotope tracer-based metabolomic approaches enable unambiguous tracking of individual atoms through compartmentalized metabolic networks directly in human subjects, which promises to decipher the complexity of the human metabolome at an unprecedented pace. This knowledge will revolutionize our understanding of complex human diseases, clinical diagnostics, as well as individualized therapeutics and drug response. In this review, we focus on the use of stable isotope tracers with metabolomics technologies for understanding metabolic network dynamics in both model systems and in clinical applications. Atom-resolved isotope tracing via the two major analytical platforms, NMR and MS, has the power to determine novel metabolic reprogramming in diseases, discover new drug targets, and facilitates ADME studies. We also illustrate new metabolic tracer-based imaging technologies, which enable direct visualization of metabolic processes in vivo. We further outline current practices and future requirements for biochemoinformatics development, which is an integral part of translating stable isotope-resolved metabolomics into clinical reality.
Collapse
Affiliation(s)
- Teresa W-M Fan
- Department of Chemistry, University of Louisville, KY 40292, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Marcucci F, Corti A. How to improve exposure of tumor cells to drugs: promoter drugs increase tumor uptake and penetration of effector drugs. Adv Drug Deliv Rev 2012; 64:53-68. [PMID: 21983328 DOI: 10.1016/j.addr.2011.09.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 08/13/2011] [Accepted: 09/20/2011] [Indexed: 12/11/2022]
Abstract
Solid tumors are characterized by an abnormal architecture and composition that limit the uptake and distribution of antitumor drugs. Over the last two decades, drugs have been identified that improve the tumor uptake and distribution of drugs that have direct antitumor effects. We propose to refer to these drugs as promoter drugs, and as effector drugs to drugs that have direct antitumor effects. Some promoter drugs have received regulatory approval, while others are in active clinical development. This review gives an overview of promoter drugs, by classifying them according to their mechanism of action: promoter drugs that modulate tumor blood flow, modify the barrier function of tumor vessels, induce tumor cell killing, and overcome stromal barriers. Eventually, we discuss those that we feel are the main conclusions to be drawn from promoter drug research that has been performed so far, and suggest areas of future investigation to improve the efficacy of promoter drugs in cancer therapy.
Collapse
Affiliation(s)
- Fabrizio Marcucci
- Centro Nazionale di Epidemiologia, Sorveglianza e Promozione della Salute (CNESPS), Istituto Superiore di Sanita' (ISS), Rome, Italy.
| | | |
Collapse
|