1
|
Ji M, Zhang Y, Duan Y, Zhao Y, Su G. Carboxymethyl-β-1,3-D-glucan (CMG)-gemcitabine conjugate improves the anticancer efficacy and alleviate the toxicity of gemcitabine (GEM). Nat Prod Res 2025:1-6. [PMID: 39789726 DOI: 10.1080/14786419.2024.2448731] [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: 08/01/2024] [Revised: 11/25/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025]
Abstract
Gemcitabine (GEM) is an antitumor drug approved by the US FDA in 1996. It is used to treat cancer and solid tumours, but its effectiveness is limited by toxicity. Carboxymethyl-β-1,3-D-glucan (CMG) is a derivative of β-glucan with improved solubility. In a study, GEM was conjugated to CMG (GG) with a stable amino acid linker to enhance efficacy and reduce toxicity. GG showed significant inhibition on LLC tumour cells with IC50 value of 2.7 compared to GEM at 0.5248, and was less toxic to normal cells. In C57BL/6 mice, GG had anti-lung cancer effects in vitro and in vivo by decreasing expression of apoptosis-related proteins. The study indicates GG's potential as an anti-tumour drug for lung cancer with reduced toxicity, paving the way for further research.
Collapse
Affiliation(s)
- Maxin Ji
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Yudong Zhang
- Institute of Functional Foods & Wines, Shenyang Pharmaceutical University, Shenyang, China
| | - Yilong Duan
- Institute of Functional Foods & Wines, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuqing Zhao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Yanbian University, Yanji, China
| | - Guangyue Su
- Institute of Functional Foods & Wines, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
2
|
Wang D, Wang X, Li Y, Wang X, Wang X, Su J, Wang A, Lv K, Liu M, Xia G. Improved Antitumor Efficiency of N4 -Tetradecyloxycarbonyl Gemcitabine-Loaded Liposomes for Pancreatic Cancer Chemotherapy. Int J Nanomedicine 2024; 19:13391-13410. [PMID: 39679246 PMCID: PMC11646436 DOI: 10.2147/ijn.s485861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Background Gemcitabine (Gem) is one of the first-line chemotherapy drugs for pancreatic cancer treatment. However, its short half-life in plasma and adverse effects limited its broader application. Methods A novel Gem derivative (N4 -tetradecyloxycarbonyl gemcitabine, tcGem) was synthesized and encapsulated into liposomes (LipotcGem) to overcome the above shortcomings. Results LipotcGem has been successfully formulated, with the average size of 115 nm, zeta potential values of -36 mV, encapsulation efficiency of up to 98%, and drug loading capacity of 8.1%. Compared to Gem, LipotcGem improved in vitro antitumor activity significantly, as evidenced by the lower IC50, the higher percentage of apoptotic cells, the stronger ability to inhibit cell migration and invasion due to the higher cellular accumulation (100 times). Additionally, the endocytosis of LipotcGem was mainly mediated by caveolae, and was then processed in the lysosome, where tcGem was released and hydrolyzed into Gem. LipotcGem inhibited tumor growth by 70% in subcutaneous xenograft model and 90% in orthotopic xenograft model, respectively. LipotcGem suppressed tumor metastasis and prolonged survival without perceptible systemic toxicity, which may be caused by the longer t1/2 in vivo (3.5 times, 5.23 vs 1.46 h) and more enrichment in tumor tissue (750 times). Conclusion LipotcGem significantly increased the anti-tumor efficiency and decreased the toxicity for chemotherapy of pancreatic cancer.
Collapse
Affiliation(s)
- Dan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Xiaobo Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Yan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Xiaowei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Xuelei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Jiayi Su
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Apeng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Kai Lv
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Mingliang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People’s Republic of China
| |
Collapse
|
3
|
Carnamucio F, Foti C, Cordaro M, Saija F, Cassone G, da Rocha SRP, Giuffrè O. Metal Complexation for the Rational Design of Gemcitabine Formulations in Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56789-56800. [PMID: 39378358 PMCID: PMC11503523 DOI: 10.1021/acsami.4c12550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024]
Abstract
Nanoformulation of chemotherapies represents a promising strategy to enhance outcomes in cancer therapy. Gemcitabine is a chemotherapeutic agent approved by the Food and Drug Administration for the treatment of various solid tumors. Nevertheless, its therapeutic effectiveness is constrained by its poor metabolic stability and pharmacokinetic profile. Nanoformulations of gemcitabine in lipid and polymer nanocarriers usually lead to poor loading capability and an inability to effectively control its release profile due to the physicochemical characteristics of the drug and matrices. Here, we propose metal-gemcitabine complexation with biorelevant metal cations as a strategy to alter the properties of gemcitabine in a noncovalent manner, paving the way for the development of novel nanoformulations. A speciation study on gemcitabine and Mn2+, Zn2+, and Ca2+ was performed with the aim of investigating the extent of the interaction between the drug and the proposed metal cations, and selecting the best conditions of temperature, pH, and drug-to-metal molar ratio that optimize such interactions. Also, a series of density functional theory calculations and spin-polarized ab initio molecular dynamics simulations were carried out to achieve insights on the atomistic modalities of these interactions. Mn2+-gemcitabine species demonstrated the ability to maintain gemcitabine's biological activity in vitro. The scientific relevance of this study lies in its potential to propose metal-gemcitabine as a valuable strategy for developing nanoformulations with optimized quality target product profiles. The work is also clinically relevant because it will lead to improved treatment outcomes, including enhanced efficacy and pharmacokinetics, decreased toxicity, and new clinical possibilities for this potent therapeutic molecule.
Collapse
Affiliation(s)
- Federica Carnamucio
- Department
of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences
- School of Pharmacy, Virginia Commonwealth
University, Richmond, Virginia 23284, United States
| | - Claudia Foti
- Dipartimento
di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Massimiliano Cordaro
- Dipartimento
di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Franz Saija
- Institute
for Chemical-Physical Processes National Research Council of Italy, Viale Ferdinando Stagno d’Alcontres,
37, 98158 Messina, Italy
| | - Giuseppe Cassone
- Institute
for Chemical-Physical Processes National Research Council of Italy, Viale Ferdinando Stagno d’Alcontres,
37, 98158 Messina, Italy
| | - Sandro R. P. da Rocha
- Department
of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences
- School of Pharmacy, Virginia Commonwealth
University, Richmond, Virginia 23284, United States
| | - Ottavia Giuffrè
- Dipartimento
di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale F. Stagno d’Alcontres
31, 98166 Messina, Italy
| |
Collapse
|
4
|
Baghdadi RA, Abdalla AN, Abourehab MA, Tulbah AS. Evaluation of the effects of a dasatinib-containing, self-emulsifying, drug delivery system on HT29 and SW420 human colorectal carcinoma cells, and MCF7 human breast adenocarcinoma cells. J Taibah Univ Med Sci 2024; 19:806-815. [PMID: 39170071 PMCID: PMC11338096 DOI: 10.1016/j.jtumed.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/13/2024] [Accepted: 07/14/2024] [Indexed: 08/23/2024] Open
Abstract
Background/Aim Dasatinib (DS), a second-generation tyrosine kinase inhibitor, functions as a multi-target small-molecule drug via targeting various tyrosine kinases involved in neoplastic cell growth. DS inhibits cancer cell replication and migration, and induces tumor cell apoptosis in a variety of solid tumors. However, it is poorly soluble in water under some pH values. Therefore, the development of a DS-containing, self-emulsifying, drug delivery system (SeDDs) could help overcome these problems in treating cancer cells. Methods Various SeDD formulations loaded with DS were developed with isopropyl myristate (oil phase), Labrafil (surfactant), and polyethylene glycol (co-surfactant). The physicochemical properties of the formulations were assessed according to droplet size, encapsulation efficiency, and in vitro drug release. The cytotoxicity of the formulations on the cancer cell lines HT29 and SW420 (human colorectal carcinoma), and MCF7 (human breast adenocarcinoma), in addition to MRC5 normal human fetal lung fibroblasts, was evaluated to assess selectivity. Results The DS-SeDD formulation showed favorable particle size, encapsulation efficiency, and in vitro drug release. The anti-cancer potency of DS-SeDDs had greater cytotoxicity effects than pure DA on the three cancer cell lines, MCF7, HT29, and SW420l. Conclusion The developed DS-SeDD formulations may potentially be an effective sustained drug delivery method for cancer therapy.
Collapse
Affiliation(s)
- Rehab A. Baghdadi
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, KSA
| | - Ashraf N. Abdalla
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, KSA
| | - Mohammed A.S. Abourehab
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, KSA
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Alaa S. Tulbah
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, KSA
| |
Collapse
|
5
|
Porter J, Noble AR, Signoret N, Fascione MA, Miller GJ. Exploring a Gemcitabine-Glucose Hybrid as a Glycoconjugate Prodrug. ACS OMEGA 2024; 9:31703-31713. [PMID: 39072123 PMCID: PMC11270703 DOI: 10.1021/acsomega.4c02417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/30/2024]
Abstract
Nucleoside analogues are established treatments for cancer and viral infection. Gemcitabine is a commonly employed nucleoside analogue displaying anticancer properties against a range of tumor types but is rapidly inactivated in vivo. Efforts to bolster its pharmaceutical profile include investigating prodrug forms. Herein, we explore the synthesis of a novel glucose-gemcitabine glycoconjugate, targeting uptake via glucose transport. We select a redox-reactive disulfide linker for conjugation of gemcitabine (through N4-cytosine) with glucose. Evaluation of this glycoconjugate reveals increased toxicity against androgen insensitive PC3 prostate cancer cells compared to LNCaP (which have lower levels of glucose transporter GLUT1). These preliminary results suggest that glycoconjugation of nucleosides may be an effective approach to targeting cells which display increased uptake and metabolism of glucose.
Collapse
Affiliation(s)
- Jack Porter
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | - Amanda R. Noble
- Hull
York Medical School, University of York, Heslington, York YO10
5DD, U.K.
| | - Nathalie Signoret
- Hull
York Medical School, University of York, Heslington, York YO10
5DD, U.K.
| | - Martin A. Fascione
- Department
of Chemistry, University of York, Heslington, York YO10
5DD, U.K.
| | - Gavin J. Miller
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| |
Collapse
|
6
|
Wang F, Liang L, Yu M, Wang W, Badar IH, Bao Y, Zhu K, Li Y, Shafi S, Li D, Diao Y, Efferth T, Xue Z, Hua X. Advances in antitumor activity and mechanism of natural steroidal saponins: A review of advances, challenges, and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155432. [PMID: 38518645 DOI: 10.1016/j.phymed.2024.155432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Cancer, the second leading cause of death worldwide following cardiovascular diseases, presents a formidable challenge in clinical settings due to the extensive toxic side effects associated with primary chemotherapy drugs employed for cancer treatment. Furthermore, the emergence of drug resistance against specific chemotherapeutic agents has further complicated the situation. Consequently, there exists an urgent imperative to investigate novel anticancer drugs. Steroidal saponins, a class of natural compounds, have demonstrated notable antitumor efficacy. Nonetheless, their translation into clinical applications has remained unrealized thus far. In light of this, we conducted a comprehensive systematic review elucidating the antitumor activity, underlying mechanisms, and inherent limitations of steroidal saponins. Additionally, we propose a series of strategic approaches and recommendations to augment the antitumor potential of steroidal saponin compounds, thereby offering prospective insights for their eventual clinical implementation. PURPOSE This review summarizes steroidal saponins' antitumor activity, mechanisms, and limitations. METHODS The data included in this review are sourced from authoritative databases such as PubMed, Web of Science, ScienceDirect, and others. RESULTS A comprehensive summary of over 40 steroidal saponin compounds with proven antitumor activity, including their applicable tumor types and structural characteristics, has been compiled. These steroidal saponins can be primarily classified into five categories: spirostanol, isospirostanol, furostanol, steroidal alkaloids, and cholestanol. The isospirostanol and cholestanol saponins are found to have more potent antitumor activity. The primary antitumor mechanisms of these saponins include tumor cell apoptosis, autophagy induction, inhibition of tumor migration, overcoming drug resistance, and cell cycle arrest. However, steroidal saponins have limitations, such as higher cytotoxicity and lower bioavailability. Furthermore, strategies to address these drawbacks have been proposed. CONCLUSION In summary, isospirostanol and cholestanol steroidal saponins demonstrate notable antitumor activity and different structural categories of steroidal saponins exhibit variations in their antitumor signaling pathways. However, the clinical application of steroidal saponins in cancer treatment still faces limitations, and further research and development are necessary to advance their potential in tumor therapy.
Collapse
Affiliation(s)
- Fengge Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR, PR China
| | - Ma Yu
- School of Life Science and Engineering, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang, 621010, Sichuan, PR China
| | - Wenjie Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Iftikhar Hussain Badar
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, PR China; Department of Meat Science and Technology, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Kai Zhu
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yanlin Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Saba Shafi
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Dangdang Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yongchao Diao
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany.
| | - Zheyong Xue
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| | - Xin Hua
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| |
Collapse
|
7
|
Handl V, Waldherr L, Arbring Sjöström T, Abrahamsson T, Seitanidou M, Erschen S, Gorischek A, Bernacka-Wojcik I, Saarela H, Tomin T, Honeder SE, Distl J, Huber W, Asslaber M, Birner-Grünberger R, Schäfer U, Berggren M, Schindl R, Patz S, Simon DT, Ghaffari-Tabrizi-Wizsy N. Continuous iontronic chemotherapy reduces brain tumor growth in embryonic avian in vivo models. J Control Release 2024; 369:668-683. [PMID: 38548064 DOI: 10.1016/j.jconrel.2024.03.044] [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: 11/15/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024]
Abstract
Local and long-lasting administration of potent chemotherapeutics is a promising therapeutic intervention to increase the efficiency of chemotherapy of hard-to-treat tumors such as the most lethal brain tumors, glioblastomas (GBM). However, despite high toxicity for GBM cells, potent chemotherapeutics such as gemcitabine (Gem) cannot be widely implemented as they do not efficiently cross the blood brain barrier (BBB). As an alternative method for continuous administration of Gem, we here operate freestanding iontronic pumps - "GemIPs" - equipped with a custom-synthesized ion exchange membrane (IEM) to treat a GBM tumor in an avian embryonic in vivo system. We compare GemIP treatment effects with a topical metronomic treatment and observe that a remarkable growth inhibition was only achieved with steady dosing via GemIPs. Daily topical drug administration (at the maximum dosage that was not lethal for the embryonic host organism) did not decrease tumor sizes, while both treatment regimes caused S-phase cell cycle arrest and apoptosis. We hypothesize that the pharmacodynamic effects generate different intratumoral drug concentration profiles for each technique, which causes this difference in outcome. We created a digital model of the experiment, which proposes a fast decay in the local drug concentration for the topical daily treatment, but a long-lasting high local concentration of Gem close to the tumor area with GemIPs. Continuous chemotherapy with iontronic devices opens new possibilities in cancer treatment: the long-lasting and highly local dosing of clinically available, potent chemotherapeutics to greatly enhance treatment efficiency without systemic side-effects. SIGNIFICANCE STATEMENT: Iontronic pumps (GemIPs) provide continuous and localized administration of the chemotherapeutic gemcitabine (Gem) for treating glioblastoma in vivo. By generating high and constant drug concentrations near the vascularized growing tumor, GemIPs offer an efficient and less harmful alternative to systemic administration. Continuous GemIP dosing resulted in remarkable growth inhibition, superior to daily topical Gem application at higher doses. Our digital modelling shows the advantages of iontronic chemotherapy in overcoming limitations of burst release and transient concentration profiles, and providing precise control over dosing profiles and local distribution. This technology holds promise for future implants, could revolutionize treatment strategies, and offers a new platform for studying the influence of timing and dosing dependencies of already-established drugs in the fight against hard-to-treat tumors.
Collapse
Affiliation(s)
- Verena Handl
- Gottfried Schatz Research Center - Medical Physics and Biophysics, Medical University of Graz, 8010 Graz, Austria
| | - Linda Waldherr
- Gottfried Schatz Research Center - Medical Physics and Biophysics, Medical University of Graz, 8010 Graz, Austria; BioTechMed-Graz, Austria, Auenbruggerplatz 30, 8036 Graz, Austria
| | - Theresia Arbring Sjöström
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 60174 Norrköping, Sweden
| | - Tobias Abrahamsson
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 60174 Norrköping, Sweden
| | - Maria Seitanidou
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 60174 Norrköping, Sweden
| | - Sabine Erschen
- Gottfried Schatz Research Center - Medical Physics and Biophysics, Medical University of Graz, 8010 Graz, Austria
| | - Astrid Gorischek
- Gottfried Schatz Research Center - Medical Physics and Biophysics, Medical University of Graz, 8010 Graz, Austria
| | - Iwona Bernacka-Wojcik
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 60174 Norrköping, Sweden
| | - Helena Saarela
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 60174 Norrköping, Sweden
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, 1060 Vienna, Austria
| | - Sophie Elisabeth Honeder
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, 1060 Vienna, Austria; Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Joachim Distl
- Gottfried Schatz Research Center - Medical Physics and Biophysics, Medical University of Graz, 8010 Graz, Austria
| | - Waltraud Huber
- Otto Loewi Research Center, Division of Immunology, Research Unit CAM Lab, Medical University of Graz, 8010 Graz, Austria
| | - Martin Asslaber
- Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Ruth Birner-Grünberger
- Institute of Chemical Technologies and Analytics, Technische Universität Wien, 1060 Vienna, Austria; Diagnostic and Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Ute Schäfer
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, 8010 Graz, Austria
| | - Magnus Berggren
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 60174 Norrköping, Sweden
| | - Rainer Schindl
- Gottfried Schatz Research Center - Medical Physics and Biophysics, Medical University of Graz, 8010 Graz, Austria; BioTechMed-Graz, Austria, Auenbruggerplatz 30, 8036 Graz, Austria.
| | - Silke Patz
- Research Unit for Experimental Neurotraumatology, Medical University of Graz, 8010 Graz, Austria.
| | - Daniel T Simon
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, 60174 Norrköping, Sweden.
| | - Nassim Ghaffari-Tabrizi-Wizsy
- Otto Loewi Research Center, Division of Immunology, Research Unit CAM Lab, Medical University of Graz, 8010 Graz, Austria.
| |
Collapse
|
8
|
Natu J, Nagaraju GP. Gemcitabine effects on tumor microenvironment of pancreatic ductal adenocarcinoma: Special focus on resistance mechanisms and metronomic therapies. Cancer Lett 2023; 573:216382. [PMID: 37666293 DOI: 10.1016/j.canlet.2023.216382] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered one of the deadliest malignancies, with dismal survival rates and extremely prevalent chemoresistance. Gemcitabine is one of the primary treatments used in treating PDACs, but its benefits are limited due to chemoresistance, which could be attributed to interactions between the tumor microenvironment (TME) and intracellular processes. In preclinical models, certain schedules of administration of gemcitabine modulate the TME in a manner that does not promote resistance. Metronomic therapy constitutes a promising strategy to overcome some barriers associated with current PDAC treatments. This review will focus on gemcitabine's mechanism in treating PDAC, combination therapies, gemcitabine's interactions with the TME, and gemcitabine in metronomic therapies.
Collapse
Affiliation(s)
- Jay Natu
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA.
| |
Collapse
|
9
|
Puris E, Fricker G, Gynther M. The Role of Solute Carrier Transporters in Efficient Anticancer Drug Delivery and Therapy. Pharmaceutics 2023; 15:pharmaceutics15020364. [PMID: 36839686 PMCID: PMC9966068 DOI: 10.3390/pharmaceutics15020364] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Transporter-mediated drug resistance is a major obstacle in anticancer drug delivery and a key reason for cancer drug therapy failure. Membrane solute carrier (SLC) transporters play a crucial role in the cellular uptake of drugs. The expression and function of the SLC transporters can be down-regulated in cancer cells, which limits the uptake of drugs into the tumor cells, resulting in the inefficiency of the drug therapy. In this review, we summarize the current understanding of low-SLC-transporter-expression-mediated drug resistance in different types of cancers. Recent advances in SLC-transporter-targeting strategies include the development of transporter-utilizing prodrugs and nanocarriers and the modulation of SLC transporter expression in cancer cells. These strategies will play an important role in the future development of anticancer drug therapies by enabling the efficient delivery of drugs into cancer cells.
Collapse
|
10
|
Design of an L-Valine-Modified Nanomicelle-Based Drug Delivery System for Overcoming Ocular Surface Barriers. Pharmaceutics 2022; 14:pharmaceutics14061277. [PMID: 35745853 PMCID: PMC9230556 DOI: 10.3390/pharmaceutics14061277] [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: 05/08/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 12/19/2022] Open
Abstract
The incidence of ocular surface disease (OSD) is increasing, with a trend towards younger ages. However, it is difficult for drugs to reach the deep layers of the cornea due to ocular surface barriers, and bioavailability is less than 5%. In this study, DSPE-PEG2000 was modified with L-valine (L-Val), and an HS15/DSPE-PEG2000-L-Val nanomicelle delivery system containing baicalin (BC) (BC@HS15/DSPE-PEG2000-L-Val) was constructed using thin-film hydration, with a high encapsulation rate, small particle size and no irritation to the ocular surface. Retention experiments on the ocular surface of rabbits and an in vivo corneal permeation test showed that, compared with the control, nanomicelles not only prolonged retention time but also enhanced the ability to deliver drugs to the deep layers of the cornea. The results of a protein inhibition and protein expression assay showed that nanomicelles could increase uptake in human corneal epithelial cells (HCEC) through energy-dependent endocytosis mediated by clathrin, caveolin and the carrier pathway mediated by PepT1 by inhibiting the overexpression of claudin-1 and ZO-1 and suppressing the expression of PepT1-induced by drug stimulation. These results indicate that BC@HS15/DSPE-PEG2000-L-Val is suitable for drug delivery to the deep layers of the ocular surface, providing a potential approach for the development of ocular drug delivery systems.
Collapse
|
11
|
Yang J, Guo H, Lei J, Zhang S, Zhang S, Bai J, Li S. Fabrication of polymer-based self-assembly nanocarriers loaded with a crizotinib and gemcitabine: potential therapeutics for the treatment of endometrial cancer. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 33:20-34. [PMID: 34602004 DOI: 10.1080/09205063.2021.1974149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Combination therapy in cancer therapy has been widely used for its positive attributes, such as minimizing the undesirable side effects of chemotherapies and enhancing the therapeutic effects on different cancers. Compared with free drugs crizotinib (CRZ) and gemcitabine (GEM), CRZ@GEM-NPs could remarkably improve the cytotoxicity for endometrial cancer (EC) cells (Ishikawa cells and KLE cells) after treatment with MTT assay. In this study, CRZ and GEM were conjugated to tri-block copolymer poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCL-PEG-PCL, known as NPs). The fabricated nanoparticles were characterized by the high-resolution transmission electron microscopy (HR-TEM), and the particles size and zeta potential were investigated by the dynamic light scattering analysis. Further, the morphological features of the EC cell lines were examined by the biochemical staining assays. Morphological changes in endometrial cells morphology revealed by nuclear fragmentation and nuclear condensation (the hallmarks of apoptosis) were noted upon treatment with CRZ@GEM-NPs to the Ishikawa and KLE cancer cells. In addition, resulting in the highest ratio of apoptosis and mitochondrial membrane potential shows the cell death through the mitochondrial membrane potential. In vivo, systemic toxicity studies showed no histological changes and substantial blood biochemical with the near-normal appearance of the organs upon treatment with CRZ@GEM-NPs. Overall, the targeted combination suitable therapeutic framework may be a promising candidate for improved EC therapy.
Collapse
Affiliation(s)
- Jiaolin Yang
- Department of Gynecology, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Hongrui Guo
- Department of Gynecology, Yuncheng Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jing Lei
- Department of Gynecology, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Sanyuan Zhang
- Department of Gynecology, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Shaoguo Zhang
- Department of Nursing Care, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jirong Bai
- Department of Gynecology, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Sufen Li
- Department of Gynecology, the First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
12
|
Churchill GC, Strupp M, Factor C, Bremova-Ertl T, Factor M, Patterson MC, Platt FM, Galione A. Acetylation turns leucine into a drug by membrane transporter switching. Sci Rep 2021; 11:15812. [PMID: 34349180 PMCID: PMC8338929 DOI: 10.1038/s41598-021-95255-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Small changes to molecules can have profound effects on their pharmacological activity as exemplified by the addition of the two-carbon acetyl group to make drugs more effective by enhancing their pharmacokinetic or pharmacodynamic properties. N-acetyl-D,L-leucine is approved in France for vertigo and its L-enantiomer is being developed as a drug for rare and common neurological disorders. However, the precise mechanistic details of how acetylation converts leucine into a drug are unknown. Here we show that acetylation of leucine switches its uptake into cells from the L-type amino acid transporter (LAT1) used by leucine to organic anion transporters (OAT1 and OAT3) and the monocarboxylate transporter type 1 (MCT1). Both the kinetics of MCT1 (lower affinity compared to LAT1) and the ubiquitous tissue expression of MCT1 make it well suited for uptake and distribution of N-acetyl-L-leucine. MCT1-mediated uptake of a N-acetyl-L-leucine as a prodrug of leucine bypasses LAT1, the rate-limiting step in activation of leucine-mediated signalling and metabolic process inside cells such as mTOR. Converting an amino acid into an anion through acetylation reveals a way for the rational design of drugs to target anion transporters.
Collapse
Affiliation(s)
- Grant C Churchill
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, UK.
| | - Michael Strupp
- Department of Neurology and German Center for Vertigo and Balance Disorders, Hospital of the Ludwig Maximilians University, Munich, Germany
| | - Cailley Factor
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, UK
| | - Tatiana Bremova-Ertl
- Department of Neurology, University Hospital Inselspital, Bern, BE, Switzerland
- Center for Rare Diseases, University Hospital Inselspital Bern, Bern, BE, Switzerland
| | - Mallory Factor
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, UK
| | - Marc C Patterson
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, UK
| |
Collapse
|
13
|
Wang Y, Wang J, Yang L, Qiu L, Hua Y, Wu S, Zeng S, Yu L, Zheng X. Epigenetic regulation of intestinal peptide transporter PEPT1 as a potential strategy for colorectal cancer sensitization. Cell Death Dis 2021; 12:532. [PMID: 34031358 PMCID: PMC8144210 DOI: 10.1038/s41419-021-03814-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022]
Abstract
Human intestinal peptide transporter PEPT1 is commonly repressed in human colorectal cancer (CRC), yet its relationship with sensitivity to the common CRC treatment ubenimex has not previously been elucidated. In this study, we confirmed PEPT1 suppression in CRC using real-time quantitative polymerase chain reaction and western blotting and then investigated the underlying epigenetic pathways involved using bisulfite sequencing, chromatin immunoprecipitation, siRNA knockdown, and reporter gene assays. We found that PEPT1 transcriptional repression was due to both DNMT1-mediated DNA methylation of the proximal promoter region and HDAC1-mediated histone deacetylation, which blocked P300-mediated H3K18/27Ac at the PEPT1 distal promoter. Finally, the effects of the epigenetic activation of PEPT1 on the CRC response to ubenimex were evaluated using sequential combination therapy of decitabine and ubenimex both in vitro and in xenografts. In conclusion, epigenetic silencing of PEPT1 due to increased DNMT1 and HDAC1 expression plays a vital role in the poor response of CRC to ubenimex.
Collapse
Affiliation(s)
- Yanhong Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Jiaqi Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Lingrong Yang
- Department of Pharmacy, Hangzhou Cancer Hospital, 310002, Hangzhou, China
| | - Liqing Qiu
- Department of Pharmacy, Hangzhou Cancer Hospital, 310002, Hangzhou, China
| | - Yuhui Hua
- Department of Pharmacy, Hangzhou Cancer Hospital, 310002, Hangzhou, China
| | - Shixiu Wu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 518116, Shenzhen, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China.
| | - Xiaoli Zheng
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 518116, Shenzhen, China.
| |
Collapse
|
14
|
Liu D, Zhang W, Liu X, Qiu R. Precise engineering of hybrid molecules-loaded macromolecular nanoparticles shows in vitro and in vivo antitumor efficacy toward the treatment of nasopharyngeal cancer cells. Drug Deliv 2021; 28:776-786. [PMID: 33866910 PMCID: PMC8079022 DOI: 10.1080/10717544.2021.1902022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Cancers continue to be the second leading cause of death worldwide. Despite the development and improvement of surgery, chemotherapy, and radiotherapy in cancer management, effective tumor ablation strategies are still in need due to high cancer patient mortality. Hence, we have established a new approach to achieve treatment-actuated modifications in a tumor microenvironment by using synergistic activity between two potential anticancer drugs. Dual drug delivery of gemcitabine (GEM) and cisplatin (PT) exhibits a great anticancer potential, as GEM enhances the effect of PT treatment of human cells by providing stability of the microenvironment. However, encapsulation of GEM and PT fanatical by methoxypoly(ethylene glycol)-block-poly(D, L-lactic acid) (PEG-PLA in termed as NPs) is incompetent owing to unsuitability between the binary Free GEM and PT core and the macromolecular system. Now, we display that PT can be prepared by hydrophobic coating of the dual drug centers with dioleoylphosphatidic acid (DOPA). The DOPA-covered PT can be co-encapsulated in PLGA NPs alongside GEM to stimulate excellent anticancer property. The occurrence of the PT suggestively enhanced the encapsulations of GEM into PLGA NPs (GEM-PT NPs). Further, the morphology of GEM NPs, PT NPs, and GEM-PT NPs and nanoparticle size was examined by transmission microscopy (TEM), respectively. Furthermore GEM-PT NPs induced significant apoptosis in human nasopharyngeal carcinoma CNE2 and SUNE1 cancer cells by in vitro. The morphological observation and apoptosis were confirmed by the various biochemical assays (AO-EB, nuclear staining, and annexin V-FITC). In a xenograft model of nasopharyngeal cancer, this nanotherapy shows a durable inhibition of tumor progression upon the administration of a tolerable dose. Our results suggest that a macromolecular hydrophobic and highly toxic drug can be rationally converted into a pharmacologically efficient and self-deliverable of nanotherapy.
Collapse
Affiliation(s)
- Dongmei Liu
- Department of Radiation Oncology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenguang Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinju Liu
- Department of Radiation Oncology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Rongliang Qiu
- Department of Radiation Oncology, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Xiao Y, Gao Y, Li F, Deng Z. Combinational dual drug delivery system to enhance the care and treatment of gastric cancer patients. Drug Deliv 2020; 27:1491-1500. [PMID: 33100060 PMCID: PMC7594745 DOI: 10.1080/10717544.2020.1822460] [Citation(s) in RCA: 9] [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] [Received: 08/05/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 01/13/2023] Open
Abstract
Gastric cancer is a frequently occurring cancer with high mortality each year worldwide. Finding new and effective therapeutic strategy against human gastric cancer is still urgently required. Hence, we have established a new method to achieve treatment-actuated modifications in a tumor microenvironment by utilizing synergistic activity between two potential anticancer drugs. Dual drug delivery of gemcitabine (GEM) and Camptothecin-11 (CPT-11) exhibits a great anti-cancer potential, as GEM enhances the effect of CPT-11 treatment of human gastric cells by providing microenvironment stability. However, encapsulation of GEM and CPT-11 obsessed by poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs) is incompetent owing to unsuitability between the binary free GEM and CPT-11 moieties and the polymeric system. Now, we display that CPT-11 can be prepared by hydrophobic covering of the drug centers with dioleoylphosphatidic acid (DOPA). The DOPA-covered CPT-11 can be co-encapsulated in PLGA NPs alongside GEM to stimulate excellent anticancer property. The occurrence of the CPT-11 suggestively enhanced the encapsulations of GEM into PLGA NPs (GEM-CPT-11 NPs). Formation of the nanocomposite (GEM-CPT-11 NPs) was confirmed by FTIR and X-ray spectroscopic techniques. Further, the morphology of GEM NPs, CPT-11 NPs, and GEM-CPT-11 NPs and NP size was examined by transmission electron microscopy (TEM), respectively. Furthermore, GEM-CPT-11 NPs induced significant apoptosis in human gastric NCI-N87 and SGC-791 cancer cells in vitro. The morphological observation and apoptosis were confirmed by the various biochemical assays (AO-EB, nuclear staining, and annexin V-FITC). In addition, evaluation of the hemolysis assay with erythrocytes of human shows excellent biocompatibility of free GEM, free CPT-11, GEM NPs, CPT-11 NPs, and GEM-CPT-11 NPs. The results suggest that GEM-CPT-11 NPs are one of the promising nursing cares for human gastric cancer therapeutic candidates worthy of further investigations.
Collapse
Affiliation(s)
- Ying Xiao
- Second Department of General Surgery, Xinxiang Central Hospital, Xinxiang, PR China
| | - Yuewen Gao
- Department of General Surgery, Rizhao People's Hospital, Rizhao, PR China
| | - Fajuan Li
- Department of General Surgery, Rizhao People's Hospital, Rizhao, PR China
| | - Zhihe Deng
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, PR China
| |
Collapse
|