1
|
Nirmala MJ, Durai L, Rao KA, Nagarajan R. Ultrasonic Nanoemulsification of Cuminum cyminum Essential Oil and Its Applications in Medicine. Int J Nanomedicine 2020; 15:795-807. [PMID: 32103937 PMCID: PMC7008196 DOI: 10.2147/ijn.s230893] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/20/2019] [Indexed: 12/29/2022] Open
Abstract
Background and Study Cumin seed oil (extracted from Cuminum cyminum) has many applications but conclusive evidence of its therapeutic uses has not been presented. This study has explored the anticancer and antibacterial properties of the seed oil. Methods The cumin nanoemulsion was prepared with Tween 80 non-ionic surfactant employing ultra-sonication technology. The anticancer activity of the nanoscale-based emulsion was evaluated through cell viability (MTT), antiproliferation evaluation through clonogenic assay, and apoptosis through Annexin V-FITC assay. Agar well diffusion was used to study the antimicrobial activity, and this was supported by membrane integrity analysis. Results A thorough study of process parameters, aimed at obtaining the optimal surface concentration and emulsification time, was completed. GC-MS data indicated cumaldehyde as a major component. The resultant droplet diameter after a sonication time of 5 min was 10.4 ± 0.5 nm. MTT assay revealed the IC50 value at 1.5 µL/mL and the early induction of apoptosis was evident. Tongue carcinoma cell line treated with cumin nanoemulsion presented a diminished colony formation. The nanoemulsion exhibited significant antibacterial activity against S. aureus. A significant cytoplasmic leakage was observed on treatment with cumin nanoemulsion. The consequences of the analysis projected cumin as a potential component for cancer therapy. Conclusion This study provides definitive evidence for cumin essential oil nanoemulsion as a legitimate plant-based medicine that can bypass the drawbacks of the present aggressive treatment of cancer, can overcome the antimicrobial resistance, and can also meet all prerequisites.
Collapse
Affiliation(s)
- M Joyce Nirmala
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai 600 036, India
| | - Latha Durai
- Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Kagitala Anvesh Rao
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai 600 036, India
| | - R Nagarajan
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai 600 036, India
| |
Collapse
|
2
|
Malik Z. Fundamentals of 5‐aminolevulinic acid photodynamic therapy and diagnosis: An overview. TRANSLATIONAL BIOPHOTONICS 2020. [DOI: 10.1002/tbio.201900022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Zvi Malik
- Faculty of Life ScienceBar‐Ilan University Ramat‐Gan Israel
| |
Collapse
|
3
|
Rabinovich OF, Guseva AV, Abramova ES. [Clinical and laboratory rationale for photodynamic therapy in patients with severe complicated oral lichen planus]. STOMATOLOGII︠A︡ 2015; 94:40-46. [PMID: 26171545 DOI: 10.17116/stomat201594240-46] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
| | - A V Guseva
- Central Research Institute of Dental and Maxillofacial
| | - E S Abramova
- Central Research Institute of Dental and Maxillofacial
| |
Collapse
|
4
|
Yang X, Li W, Palasuberniam P, Myers KA, Wang C, Chen B. Effects of Silencing Heme Biosynthesis Enzymes on 5-Aminolevulinic Acid-mediated Protoporphyrin IX Fluorescence and Photodynamic Therapy. Photochem Photobiol 2015; 91:923-30. [PMID: 25809721 DOI: 10.1111/php.12454] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/06/2015] [Indexed: 01/31/2023]
Abstract
Aminolevulinic acid (ALA)-mediated protoporphyrin IX (PpIX) production is being explored for tumor fluorescence imaging and photodynamic therapy (PDT). As a prodrug, ALA is converted in heme biosynthesis pathway to PpIX with fluorescent and photosensitizing properties. To better understand the role of heme biosynthesis enzymes in ALA-mediated PpIX fluorescence and PDT efficacy, we used lentiviral shRNA to silence the expression of porphobilinogen synthase (PBGS), porphobilinogen deaminase (PBGD) and ferrochelatase (FECH) in SkBr3 human breast cancer cells. PBGS and PBGD are the first two cytosolic enzymes involved in PpIX biosynthesis, and FECH is the enzyme responsible for converting PpIX to heme. PpIX fluorescence was examined by flow cytometry and confocal fluorescence microscopy. Cytotoxicity was assessed after ALA-mediated PDT. Silencing PBGS or PBGD significantly reduced ALA-stimulated PpIX fluorescence, whereas silencing FECH elevated basal and ALA-stimulated PpIX fluorescence. However, compared with vector control cells, the ratio of ALA-stimulated fluorescence to basal fluorescence without ALA was significantly reduced in all knockdown cell lines. PBGS or PBGD knockdown cells exhibited significant resistance to ALA-PDT, while increased sensitivity to ALA-PDT was found in FECH knockdown cells. These results demonstrate the importance of PBGS, PBGD and FECH in ALA-mediated PpIX fluorescence and PDT efficacy.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA
| | - Weihua Li
- Key Laboratory of Tianjin Radiation and Molecular Nuclear Medicine; Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Pratheeba Palasuberniam
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA
| | - Kenneth A Myers
- Department of Biological Sciences, Misher College of Arts & Sciences, University of the Sciences, Philadelphia, PA
| | - Chenguang Wang
- Key Laboratory of Tianjin Radiation and Molecular Nuclear Medicine; Institute of Radiation Medicine, Peking Union Medical College & Chinese Academy of Medical Sciences, Tianjin, China
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA
| |
Collapse
|
5
|
Suzuki C, Tsuji AB, Kato K, Kikuchi T, Sudo H, Okada M, Sugyo A, Zhang MR, Arano Y, Saga T. Preclinical characterization of 5-amino-4-oxo-[6-11C]hexanoic acid as an imaging probe to estimate protoporphyrin IX accumulation induced by exogenous aminolevulinic acid. J Nucl Med 2014; 55:1671-7. [PMID: 25125482 DOI: 10.2967/jnumed.114.145086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Preoperative noninvasive imaging to estimate the quantity and spatial distribution of protoporphyrin IX (PpIX) accumulation in tumors induced by 5-aminolevulinic acid (ALA) administration is expected to improve the efficacy of ALA-based fluorescence-guided resection and photo- and sonodynamic therapies. PpIX synthesis from exogenous ALA has been reported to be regulated by ALA influx or ALA dehydratase (ALAD) activity, which catalyzes the first step of the synthesis. In this study, we characterized the properties of a (11)C-labeled ALA analog, 5-amino-4-oxo-[6-(11)C]hexanoic acid ((11)C-MALA), as a PET tracer to estimate PpIX accumulation. METHODS In vitro uptake of (11)C-MALA and (3)H-ALA was determined in 5 tumor cell lines after 10-min incubation with each tracer at 37°C. The expression levels of ALAD were determined by Western blot analysis. In vivo distribution and dynamic PET studies were conducted in tumor-bearing mice. In vitro and in vivo accumulation of ALA-induced PpIX was determined by measuring fluorescence in extracts of cells or tumors. RESULTS In vitro uptake of (11)C-MALA in 5 tumor cell lines was correlated with ALAD expression levels and PpIX accumulation. In vivo biodistribution and dynamic PET studies showed that (11)C-MALA was rapidly incorporated into tumors, and the tumor-to-muscle ratio of (11)C-MALA at 1 min after injection was significantly correlated with that of (3)H-ALA. (11)C-MALA in tumors was continuously decreased thereafter, and the elimination rate of (11)C-MALA from AsPC-1 tumors with the highest ALAD expression level was slower than from other tumors with lower expression levels. These results suggest that the influx and intracellular retention of (11)C-MALA reflect ALA influx and ALAD expression levels, respectively. Tumor accumulation of (11)C-MALA at 60 min after injection was strongly correlated with PpIX accumulation in tumor tissues. CONCLUSION (11)C-MALA PET has the potential to noninvasively estimate the quantitative and spatial accumulation of exogenous ALA-induced PpIX.
Collapse
Affiliation(s)
- Chie Suzuki
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Atsushi B Tsuji
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Koichi Kato
- Molecular Probe Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan; and Department of Integrative Brain Imaging, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tatsuya Kikuchi
- Molecular Probe Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan; and
| | - Hitomi Sudo
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Maki Okada
- Molecular Probe Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan; and
| | - Aya Sugyo
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Ming-Rong Zhang
- Molecular Probe Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan; and
| | - Yasushi Arano
- Department of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Tsuneo Saga
- Diagnostic Imaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| |
Collapse
|
6
|
Kiesslich T, Helander L, Illig R, Oberdanner C, Wagner A, Lettner H, Jakab M, Plaetzer K. Real-time analysis of endogenous protoporphyrin IX fluorescence from δ-aminolevulinic acid and its derivatives reveals distinct time- and dose-dependent characteristics in vitro. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:085007. [PMID: 25117078 DOI: 10.1117/1.jbo.19.8.085007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 07/14/2014] [Indexed: 06/03/2023]
Abstract
Photodynamic therapy (PDT) and photodiagnosis based on the intracellular production of the photosensitizer protoporphyrin IX (PPIX) by administration of its metabolic precursor -aminolevulinic acid (ALA) achieved their breakthrough upon the clinical approval of MAL (ALA methyl ester) and HAL (ALA hexyl ester). For newly developed ALA derivatives or application in new tumor types, in vitro determination of PPIX formation involves multiparametric experiments covering variable pro-drug concentrations, medium composition, time points of analysis, and cell type(s). This study uses a fluorescence microplate reader with a built-in temperature and atmosphere control to investigate the high-resolution long-term kinetics (72 h) of cellular PPIX fueled by administration of either ALA, MAL, or HAL for each 10 different concentrations. For simultaneous proliferation correction, A431 cells were stably transfected with green fluorescent protein. The results indicate that the peak PPIX level is a function of both, incubation concentration and period: maximal PPIX is generated with 1 to 2-mM ALA/MAL or 0.125-mM HAL; also, the PPIX peak shifts to longer incubation periods with increasing pro-drug concentrations. The results underline the need for detailed temporal analysis of PPIX formation to optimize ALA (derivative)-based PDT or photodiagnosis and highlight the value of environment-controlled microplate readers for automated in vitro analysis.
Collapse
Affiliation(s)
- Tobias Kiesslich
- Paracelsus Medical University, Institute of Physiology and Pathophysiology, Strubergasse 21, Salzburg A-5020, AustriabParacelsus Medical University/Salzburger Landeskliniken, Department of Internal Medicine I, Muellner Hauptstrasse 48, Salzburg A-5020, Au
| | - Linda Helander
- Norwegian University of Science and Technology, Department of Cancer Research and Molecular Medicine, Erling Skjalgssons gate 1, N-7491 Trondheim, Norway
| | - Romana Illig
- Paracelsus Medical University/Salzburger Landeskliniken, Institute of Pathology, Muellner Hauptstrasse 48, Salzburg A-5020, Austria
| | | | - Andrej Wagner
- Paracelsus Medical University/Salzburger Landeskliniken, Department of Internal Medicine I, Muellner Hauptstrasse 48, Salzburg A-5020, Austria
| | - Herbert Lettner
- University of Salzburg, Department of Materials Science and Physics, Division of Physics and Biophysics, Hellbrunnerstraße 34, Salzburg A-5020, Austria
| | - Martin Jakab
- Paracelsus Medical University, Institute of Physiology and Pathophysiology, Strubergasse 21, Salzburg A-5020, Austria
| | - Kristjan Plaetzer
- University of Salzburg, Department of Materials Science and Physics, Laboratory of Photodynamic Inactivation of Microorganisms, Hellbrunnerstraße 34, Salzburg A-5020, Austria
| |
Collapse
|
7
|
Suzuki C, Kato K, Tsuji AB, Kikuchi T, Zhang MR, Arano Y, Saga T. Synthesis and in vitro cellular uptake of 11C-labeled 5-aminolevulinic acid derivative to estimate the induced cellular accumulation of protoporphyrin IX. Bioorg Med Chem Lett 2013; 23:4567-70. [DOI: 10.1016/j.bmcl.2013.06.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 06/07/2013] [Accepted: 06/11/2013] [Indexed: 11/27/2022]
|
8
|
Sawamoto M, Imai T, Umeda M, Fukuda K, Kataoka T, Taketani S. The p53-dependent expression of frataxin controls 5-aminolevulinic acid-induced accumulation of protoporphyrin IX and photo-damage in cancerous cells. Photochem Photobiol 2013; 89:163-72. [PMID: 22862424 DOI: 10.1111/j.1751-1097.2012.01215.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 07/25/2012] [Indexed: 11/29/2022]
Abstract
Mitochondrial frataxin is involved in various functions such as iron homeostasis, iron-sulfur cluster biogenesis, the protection from oxidative stress and apoptosis and acts as a tumor suppressor protein. We now show that the expression of frataxin is stimulated in a p53-dependent manner and prove that frataxin is a direct p53 target gene by showing that the p53-responsive element in the promoter of the mouse frataxin gene is bound by p53. The bacterial expression of human frataxin stimulated maturation of human ferrochelatase, which catalyzes the insertion of iron into protoporphyrin at the last step of heme biosynthesis. Overexpression of frataxin in human cancer A431 and HeLa cells lowered 5-aminolevulinic acid(ALA)-induced accumulation of protoporphyrin and induced resistance to ALA-induced photo-damage, whereas p53 silencing with siRNA in non tumor HEK293T cells down-regulated the expression of frataxin and increased the accumulation of protoporphyrin. Thus, the decrease of the expression of frataxin unregulated by p53 in tumor cells enhances ALA-induced photo-damage, by down-regulation of mitochondrial functions.
Collapse
Affiliation(s)
- Mari Sawamoto
- Department of Biotechnology, Kyoto Institute of Technology, Kyoto, Japan
| | | | | | | | | | | |
Collapse
|
9
|
Stockert JC, Blázquez-Castro A, Cañete M, Horobin RW, Villanueva A. MTT assay for cell viability: Intracellular localization of the formazan product is in lipid droplets. Acta Histochem 2012; 114:785-96. [PMID: 22341561 DOI: 10.1016/j.acthis.2012.01.006] [Citation(s) in RCA: 372] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 01/20/2012] [Accepted: 01/22/2012] [Indexed: 01/01/2023]
Abstract
Although MTT is widely used to assess cytotoxicity and cell viability, the precise localization of its reduced formazan product is still unclear. In the present study the localization of MTT formazan was studied by direct microscopic observation of living HeLa cells and by colocalization analysis with organelle-selective fluorescent probes. MTT formazan granules did not colocalize with mitochondria as revealed by rhodamine 123 labeling or autofluorescence. Likewise, no colocalization was observed between MTT formazan granules and lysosomes labeled by neutral red. Taking into account the lipophilic character and lipid solubility of MTT formazan, an evaluation of the MTT reaction was performed after treatment of cells with sunflower oil emulsions to induce a massive occurrence of lipid droplets. Under this condition, lipid droplets revealed a large amount of MTT formazan deposits. Kinetic studies on the viability of MTT-treated cells showed no harmful effects at short times. Quantitative structure-activity relations (QSAR) models were used to predict and explain the localization of both the MTT tetrazolium salt and its formazan product. These predictions were in agreement with experimental observations on the accumulation of MTT formazan product in lipid droplets.
Collapse
Affiliation(s)
- Juan C Stockert
- Department of Biology, Faculty of Sciences, Autonomous University of Madrid, Spain.
| | | | | | | | | |
Collapse
|
10
|
Anand S, Ortel BJ, Pereira SP, Hasan T, Maytin EV. Biomodulatory approaches to photodynamic therapy for solid tumors. Cancer Lett 2012; 326:8-16. [PMID: 22842096 DOI: 10.1016/j.canlet.2012.07.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 07/19/2012] [Accepted: 07/19/2012] [Indexed: 12/12/2022]
Abstract
Photodynamic Therapy (PDT) uses a photosensitizing drug in combination with visible light to kill cancer cells. PDT has an advantage over surgery or ionizing radiation because PDT can eliminate tumors without causing fibrosis or scarring. Disadvantages include the dual need for drug and light, and a generally lower efficacy for PDT vs. surgery. This minireview describes basic principles of PDT, photosensitizers available, and aspects of tumor biology that may provide further opportunities for treatment optimization. An emerging biomodulatory approach, using methotrexate or Vitamin D in combination with aminolevulinate-based PDT, is described. Finally, current clinical uses of PDT for solid malignancies are reviewed.
Collapse
Affiliation(s)
- Sanjay Anand
- Department of Dermatology, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
11
|
Ohgari Y, Miyata Y, Miyagi T, Gotoh S, Ohta T, Kataoka T, Furuyama K, Taketani S. Roles of porphyrin and iron metabolisms in the δ-aminolevulinic acid (ALA)-induced accumulation of protoporphyrin and photodamage of tumor cells. Photochem Photobiol 2011; 87:1138-45. [PMID: 21668870 DOI: 10.1111/j.1751-1097.2011.00950.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
δ-Aminolevulinic acid (ALA)-induced porphyrin accumulation is widely used in the treatment of cancer, as photodynamic therapy. To clarify the mechanisms of the tumor-preferential accumulation of protoporphyrin, we examined the effect of the expression of heme-biosynthetic and -degradative enzymes on the ALA-induced accumulation of protoporphyrin as well as photodamage. The transient expression of heme-biosynthetic enzymes in HeLa cells caused variations of the ALA-induced accumulation of protoporphyrin. When ALA-treated cells were exposed to white light, the extent of photodamage of the cells was dependent on the accumulation of protoporphyrin. The decrease of the accumulation of protoporphyrin was observed in the cells treated with inducers of heme oxygenase (HO)-1. The ALA-dependent accumulation of protoporphyrin was decreased in HeLa cells by transfection with HO-1 and HO-2 cDNA. Conversely, knockdown of HO-1/-2 with siRNAs enhanced the ALA-induced protoporphyrin accumulation and photodamage. The ALA effect was decreased with HeLa cells expressing mitoferrin-2, a mitochondrial iron transporter, whereas it was enhanced by the mitoferrin-2 siRNA transfection. These results indicated that not only the production of porphyrin intermediates but also the reuse of iron from heme and mitochondrial iron utilization control the ALA-induced accumulation of protoporphyrin in cancerous cells.
Collapse
Affiliation(s)
- Yoshiko Ohgari
- Department of Biotechnology, Kyoto Institute of Technology, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Barth BM, I Altinoğlu E, Shanmugavelandy SS, Kaiser JM, Crespo-Gonzalez D, DiVittore NA, McGovern C, Goff TM, Keasey NR, Adair JH, Loughran TP, Claxton DF, Kester M. Targeted indocyanine-green-loaded calcium phosphosilicate nanoparticles for in vivo photodynamic therapy of leukemia. ACS NANO 2011; 5:5325-5337. [PMID: 21675727 DOI: 10.1021/nn2005766] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Leukemia is one of the most common and aggressive adult cancers, as well as the most prevalent childhood cancer. Leukemia is a cancer of the hematological system and can be divided into a diversity of unique malignancies based on the onset of the disease as well as the specific cell lineages involved. Cancer stem cells, including recently identified leukemia stem cells (LSCs), are hypothesized to be responsible for cancer development, relapse, and resistance to treatment, and new therapeutics targeting these cellular populations are urgently needed. Nontoxic and nonaggregating calcium phosphosilicate nanoparticles (CPSNPs) encapsulating the near-infrared fluoroprobe indocyanine green (ICG) were recently developed for diagnostic imaging and drug delivery as well as for photodynamic therapy (PDT) of solid tumors. Prior studies revealed that specific targeting of CPSNPs allowed for enhanced accumulation within breast cancer tumors, via CD71 targeting, or pancreatic cancer tumors, via gastrin receptor targeting. In the present study, ICG-loaded CPSNPs were evaluated as photosensitizers for PDT of leukemia. Using a novel bioconjugation approach to specifically target CD117 or CD96, surface features enhanced on leukemia stem cells, in vitro ICG-CPSNP PDT of a murine leukemia cell line and human leukemia samples were dramatically improved. Furthermore, the in vivo efficacy of PDT was dramatically enhanced in a murine leukemia model by utilizing CD117-targeted ICG-CPSNPs, resulting in 29% disease-free survival. Altogether, this study demonstrates that leukemia-targeted ICG-loaded CPSNPs offer the promise to effectively treat relapsing and multidrug-resistant leukemia and to improve the life of leukemia patients.
Collapse
Affiliation(s)
- Brian M Barth
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Schauder A, Feuerstein T, Malik Z. The centrality of PBGD expression levels on ALA-PDT efficacy. Photochem Photobiol Sci 2011; 10:1310-7. [PMID: 21655622 DOI: 10.1039/c1pp05085k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Successful 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) is dependent on efficient porphyrin synthesis in the inflicted cancer tissue, which is regulated by several enzymes. Irradiation of the tumor excites the light-sensitive porphyrins and results in ROS production and cell death. In this study we investigated the effect of the expression levels of two main enzymes in heme biosynthesis, ALA dehydratase (ALAD) and porphobilinogen deaminase (PBGD), on the capacity of K562 cells to undergo cell death following ALA-PDT. We manipulated PBGD and ALAD expression levels by shRNAs and PBGD overexpressing plasmid. PBGD down-regulation induced an elevation in ALAD activity, while overexpression of PBGD reduced ALAD activity, indicating a novel regulation feedback of PBGD on ALAD activity. This feedback mechanism enabled partial PpIX synthesis under PBGD silencing, whereas ALAD silencing reduced PpIX production to a minimum. ALA-PDT efficacy was directly correlated to PpIX levels. Thus, only ALAD-silenced cells were not affected by ALA+ irradiation, while following PBGD silencing, the accumulated PpIX, though decreased, was sufficient for successful ALA-PDT. The alterations in ALAD activity level initiated by changes in PBGD expression indicates PBGD's central role in heme synthesis. This enables efficient ALA-PDT, even when PBGD is not fully active. Conversely, ALAD loss resulted in reduced PpIX synthesis and consequently failure in ALA-PDT, due to the absence of compensation mechanism for ALAD.
Collapse
Affiliation(s)
- Avital Schauder
- Microscopy Unit, Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, 52900, Israel.
| | | | | |
Collapse
|
14
|
Lawrence SH, Selwood T, Jaffe EK. Diverse clinical compounds alter the quaternary structure and inhibit the activity of an essential enzyme. ChemMedChem 2011; 6:1067-73. [PMID: 21506274 PMCID: PMC3236527 DOI: 10.1002/cmdc.201100009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 02/24/2011] [Indexed: 11/10/2022]
Abstract
An in vitro evaluation of the Johns Hopkins Clinical Compound Library demonstrates that certain drugs can alter the quaternary structure of an essential human protein. Human porphobilinogen synthase (HsPBGS) is an essential enzyme involved in heme biosynthesis; it exists as an equilibrium of high-activity octamers, low-activity hexamers, and alternate dimer configurations that dictate the stoichiometry and architecture of further assembly. Decreased HsPBGS activity is implicated in toxicities associated with lead poisoning and 5-aminolevulinate dehydratase (ALAD) porphyria, the latter of which involves hexamer-favoring HsPBGS variants. A medium-throughput native PAGE mobility-shift screen coupled with evaluation of hits as HsPBGS inhibitors revealed 12 drugs that stabilize the HsPBGS hexamer and inhibit HsPBGS activity in vitro. A detailed characterization of these effects is presented. Drug inhibition of HsPBGS in vivo by inducing hexamer formation would constitute an unprecedented mechanism for side effects. We suggest that small-molecule perturbation of quaternary structure equilibria be considered as a general mechanism for drug action and side effects.
Collapse
Affiliation(s)
- Sarah H. Lawrence
- Developmental Therapeutics, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111
| | - Trevor Selwood
- Developmental Therapeutics, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111
| | - Eileen K. Jaffe
- Developmental Therapeutics, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111
| |
Collapse
|
15
|
Wachowska M, Muchowicz A, Firczuk M, Gabrysiak M, Winiarska M, Wańczyk M, Bojarczuk K, Golab J. Aminolevulinic Acid (ALA) as a Prodrug in Photodynamic Therapy of Cancer. Molecules 2011. [PMCID: PMC6263343 DOI: 10.3390/molecules16054140] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Aminolevulinic acid (ALA) is an endogenous metabolite normally formed in the mitochondria from succinyl-CoA and glycine. Conjugation of eight ALA molecules yields protoporphyrin IX (PpIX) and finally leads to formation of heme. Conversion of PpIX to its downstream substrates requires the activity of a rate-limiting enzyme ferrochelatase. When ALA is administered externally the abundantly produced PpIX cannot be quickly converted to its final product - heme by ferrochelatase and therefore accumulates within cells. Since PpIX is a potent photosensitizer this metabolic pathway can be exploited in photodynamic therapy (PDT). This is an already approved therapeutic strategy making ALA one of the most successful prodrugs used in cancer treatment.
Collapse
Affiliation(s)
- Małgorzata Wachowska
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
| | - Angelika Muchowicz
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
| | - Małgorzata Firczuk
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
| | - Magdalena Gabrysiak
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
| | - Magdalena Winiarska
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
| | - Małgorzata Wańczyk
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
| | - Kamil Bojarczuk
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Centre of Biostructure Research, Medical University of Warsaw, Banacha 1A F Building, 02-097 Warsaw, Poland
- Department III, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
- Author to whom correspondence should be addressed; E-Mail: ; Tel. +48-22-5992199; Fax: +48-22-5992194
| |
Collapse
|
16
|
Multifunctional 5-aminolevulinic acid prodrugs activating diverse cell-death pathways. Invest New Drugs 2011; 30:1028-38. [DOI: 10.1007/s10637-011-9669-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 04/04/2011] [Indexed: 10/18/2022]
|