1
|
Mahgoub TM, Jordan EJ, Mahdi AF, Oettl V, Huefner S, O'Donovan N, Crown J, Collins DM. Evaluation of ABT-751, a novel anti-mitotic agent able to overcome multi-drug resistance, in melanoma cells. Cancer Chemother Pharmacol 2024; 93:427-437. [PMID: 38226983 PMCID: PMC11043045 DOI: 10.1007/s00280-023-04624-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/20/2023] [Indexed: 01/17/2024]
Abstract
PURPOSE Drug efflux transporter associated multi-drug resistance (MDR) is a potential limitation in the use of taxane chemotherapies for the treatment of metastatic melanoma. ABT-751 is an orally bioavailable microtubule-binding agent capable of overcoming MDR and proposed as an alternative to taxane-based therapies. METHODS This study compares ABT-751 to taxanes in vitro, utilizing seven melanoma cell line models, publicly available gene expression and drug sensitivity databases, a lung cancer cell line model of MDR drug efflux transporter overexpression (DLKP-A), and drug efflux transporter ATPase assays. RESULTS Melanoma cell lines exhibit a low but variable protein and RNA expression of drug efflux transporters P-gp, BCRP, and MDR3. Expression of P-gp and MDR3 correlates with sensitivity to taxanes, but not to ABT-751. The anti-proliferative IC50 profile of ABT-751 was higher than the taxanes docetaxel and paclitaxel in the melanoma cell line panel, but fell within clinically achievable parameters. ABT-751 IC50 was not impacted by P-gp-overexpression in DKLP-A cells, which display strong resistance to the P-gp substrate taxanes compared to DLKP parental controls. The addition of ABT-751 to paclitaxel treatment significantly decreased cell proliferation, suggesting some reversal of MDR. ATPase activity assays suggest that ABT-751 is a potential BCRP substrate, with the ability to inhibit P-gp ATPase activity. CONCLUSION Our study confirms that ABT-751 is active against melanoma cell lines and models of MDR at physiologically relevant concentrations, it inhibits P-gp ATPase activity, and it may be a BCRP and/or MDR3 substrate. ABT-751 warrants further investigation alone or in tandem with other drug efflux transporter inhibitors for hard-to-treat MDR melanoma.
Collapse
Affiliation(s)
- Thamir M Mahgoub
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Emmet J Jordan
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Amira F Mahdi
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
- School of Medicine, University of Limerick, Limerick, Ireland
| | - Veronika Oettl
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Stefanie Huefner
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Norma O'Donovan
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - John Crown
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
- Department of Medical Oncology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - Denis M Collins
- Cancer Biotherapeutics Research Group, School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
2
|
Dana PM, Hallajzadeh J, Asemi Z, Mansournia MA, Yousefi B. Advances in Chitosan-based Drug Delivery Systems in Melanoma: A Narrative Review. Curr Med Chem 2024; 31:3488-3501. [PMID: 37202890 DOI: 10.2174/0929867330666230518143654] [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: 12/04/2022] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/20/2023]
Abstract
Melanoma accounts for the minority of skin cancer cases. However, it has the highest mortality rate among the subtypes of skin cancer. At the early stages of the disease, patients show a good prognosis after the surgery, but developing metastases leads to a remarkable drop in patients' 5-year survival rate. Despite the advances made in the therapeutic approaches to this disease, melanoma treatment is still facing several obstacles. Systemic toxicity, water insolubility, instability, lack of proper biodistribution, inadequate cellular penetration, and rapid clearance are some of the challenges that should be addressed in the field of melanoma treatment. While various delivery systems have been developed to circumvent these challenges, chitosan-based delivery platforms have indicated significant success. Chitosan that is produced by the deacetylation of chitin can be formulated into different materials (e.g., nanoparticle, film, and hydrogel) due to its characteristics. Both in vitro and in vivo studies have reported that chitosan-based materials can be used in drug delivery systems while offering a solution for the common problems in this area, such as enhancing biodistribution and skin penetration as well as the sustained release of the drugs. Herein, we reviewed the studies concerning the role of chitosan as a drug delivery system in melanoma and discussed how these drug systems are used for delivering chemotherapeutic drugs (e.g., doxorubicin and paclitaxel), genes (e.g., TRAIL), and RNAs (e.g., miRNA199a and STAT3 siRNA) successfully. Furthermore, we take a look into the role of chitosan-based nanoparticles in neutron capture therapy.
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Nkune NW, Abrahamse H. The Efficacy of Zinc Phthalocyanine Nanoconjugate on Melanoma Cells Grown as Three-Dimensional Multicellular Tumour Spheroids. Pharmaceutics 2023; 15:2264. [PMID: 37765232 PMCID: PMC10535874 DOI: 10.3390/pharmaceutics15092264] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Melanoma remains a major public health concern that is highly resistant to standard therapeutic approaches. Photodynamic therapy (PDT) is an underutilised cancer therapy with an increased potency and negligible side effects, and it is non-invasive compared to traditional treatment modalities. Three-dimensional multicellular tumour spheroids (MCTS) closely resemble in vivo avascular tumour features, allowing for the more efficient and precise screening of novel anticancer agents with various treatment combinations. In this study, we utilised A375 human melanoma spheroids to screen the phototoxic effect of zinc phthalocyanine tetrasulfonate (ZnPcS4) conjugated to gold nanoparticles (AuNP). The nanoconjugate was synthesised and characterised using ultraviolet-visible spectroscopy, a high-resolution transmission electron microscope (TEM), dynamic light scattering (DLS), and zeta potential (ZP). The phototoxicity of the nanoconjugate was tested on the A375 MCTS using PDT at a fluency of 10 J/cm2. After 24 h, the cellular responses were evaluated via microscopy, an MTT viability assay, an ATP luminescence assay, and cell death induction using annexin propidium iodide. The MTT viability assay demonstrated that the photoactivated ZnPcS4, at a concentration of 12.73 µM, caused an approximately 50% reduction in the cell viability of the spheroids. When conjugated to AuNPs, the latter significantly increased the cellular uptake and cytotoxicity in the melanoma spheroids via the induction of apoptosis. This novel Zinc Phthalocyanine Nanoconjugate shows promise as a more effective PDT treatment modality.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa;
| |
Collapse
|
4
|
Prajapat VM, Mahajan S, Paul PG, Aalhate M, Mehandole A, Madan J, Dua K, Chellappan DK, Singh SK, Singh PK. Nanomedicine: A pragmatic approach for tackling melanoma skin cancer. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
5
|
Jain N, Shahrukh S, Famta P, Shah S, Vambhurkar G, Khatri DK, Singh SB, Srivastava S. Immune cell-camouflaged surface-engineered nanotherapeutics for cancer management. Acta Biomater 2023; 155:57-79. [PMID: 36347447 DOI: 10.1016/j.actbio.2022.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
Abstract
Nanocarriers (NCs) have shown potential in delivering hydrophobic cytotoxic drugs and tumor-specific targeting. However, the inability to penetrate the tumor microenvironment and entrapment by macrophages has limited their clinical translation. Various cell-based drug delivery systems have been explored for their ability to improve circulation half-life and tumor accumulation capabilities. Tumors are characterized by high inflammation, which aids in tumor progression and metastasis. Immune cells show natural tumor tropism and penetration inside the tumor microenvironment (TME) and are a topic of great interest in cancer drug delivery. However, the TME is immunosuppressive and can polarize immune cells to pro-tumor. Thus, the use of immune cell membrane-coated NCs has gained popularity. Such carriers display immune cell-specific surface receptors for tumor-specific accumulation but lack cell machinery. The lack of immune cell machinery makes them unaffected by the immunosuppressive TME, meanwhile maintaining the inherent tumor tropism. In this review, we discuss the molecular mechanism behind the movement of various immune cells toward TME, the preparation and characterization of membrane-coated NCs, and the efficacy of immune cell-mimicking NCs in tumor therapy. Regulatory guidelines and the bottlenecks in clinical translation are also highlighted. STATEMENT OF SIGNIFICANCE: Nanocarriers have been explored for the site-specific delivery of chemotherapeutics. However, low systemic circulation half-life, extensive entrapment by macrophages, and poor accumulation inside the tumor microenvironment prevent the clinical translation of conventional nanotherapeutics. Immune cells possess the natural tropism towards the tumor along the chemokine gradient. Hence, coating the nanocarriers with immune cell-derived membranes can improve the accumulation of nanocarriers inside the tumor. Moreover, coating with membranes derived autologous immune cells will prevent engulfment by the macrophages.
Collapse
Affiliation(s)
- Naitik Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Syed Shahrukh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Ganesh Vambhurkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India.
| |
Collapse
|
6
|
Fard GH, Moinipoor Z, Anastasova-Ivanova S, Iqbal HM, Dwek MV, Getting S, Keshavarz T. Development of chitosan, pullulan, and alginate based drug-loaded nano-emulsions as a potential malignant melanoma delivery platform. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2022. [DOI: 10.1016/j.carpta.2022.100250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
7
|
Barjasteh M, Vossoughi M, Bagherzadeh M, Pooshang Bagheri K. Green synthesis of PEG-coated MIL-100(Fe) for controlled release of dacarbazine and its anticancer potential against human melanoma cells. Int J Pharm 2022; 618:121647. [PMID: 35288221 DOI: 10.1016/j.ijpharm.2022.121647] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
In this study, the potential of using MIL-100(Fe) metal-organic framework (MOF) for loading and controlling the release of dacarbazine (DTIC) was evaluated for in vitro treatment of melanoma. The drug loading was performed during the green synthesis of MIL-100(Fe) in an aqueous media without using any harmful solvents, to obtain MIL-DTIC. The surface of this structure was then coated with polyethylene glycol (PEG) in the same aqueous solution to synthesize MIL-DTIC-PEG. The synthesized samples were characterized using various methods. Their release profile was studied in phosphate-buffered saline (PBS) and simulated cutaneous medium (SCM). The cytotoxicity of DTIC and its nano-MOF formulation were investigated against melanoma A375 cell lines. The results revealed that the PEG coating (PEGylation) changed the surface charge of MOF from -2.8 ± 0.9 mV to -42.8 ± 1.2 mV, which can contribute to the colloidal stability of MOF. The PEGylation showed a significant effect on controlled drug release, especially in SCM, which increases the complete release time from 60 h to 12 days. Moreover, both of the drug-containing MOFs showed more toxicity than DTIC and unloaded MOFs, confirming that the cumulative release of drug and better cellular uptake of NPs lead to increased toxicity.
Collapse
Affiliation(s)
- Mahdi Barjasteh
- Institute for Nano-science and Nanotechnology, Sharif University of Technology, Tehran, Iran.
| | - Manouchehr Vossoughi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran.
| | | | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Lab., Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
8
|
Al-Zoubi MS, Al-Zoubi RM. Nanomedicine Tactics in Cancer Treatment: Challenge and Hope. Crit Rev Oncol Hematol 2022; 174:103677. [PMID: 35385774 DOI: 10.1016/j.critrevonc.2022.103677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 10/18/2022] Open
|
9
|
Sallam MA, Wyatt Shields Iv C, Prakash S, Kim J, Pan DC, Mitragotri S. A dual macrophage polarizer conjugate for synergistic melanoma therapy. J Control Release 2021; 335:333-344. [PMID: 34048840 DOI: 10.1016/j.jconrel.2021.05.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/08/2021] [Accepted: 05/23/2021] [Indexed: 12/16/2022]
Abstract
Tumor associated macrophages (TAMs) play a paradoxical role in the fate of aggressive tumors like melanoma. Immune modulation of TAMs from the tumor-permissive M2 phenotype to antitumoral M1 phenotype is an emerging attractive approach in melanoma therapy. Resiquimod is a TLR7/8 agonist that shifts the polarization of macrophages towards M1 phenotype. Bexarotene (BEX) is a retinoid that induce the expression of phagocytic receptors in macrophages besides its ability to downregulate the M2 polarization. However, the clinical use of both agents is hindered by poor pharmacokinetic properties. Here, for the first time we repurposed BEX based on its immunomodulatory properties and combined it with RES by designing hyaluronic acid (HA) conjugates of both drugs that act synergistically as a dual macrophage polarizer to promote the M1 phenotype and suppress the M2 phenotype. This combination enhanced the macrophage secretion of proinflammatory cytokines (IL-6 and TNF-α), while suppressing the production of tumor promoting cytokine CCL22. It enhanced the macrophage phagocytic ability and showed superior inhibitory effects against B16F10 cells. In vivo studies on a mouse melanoma model confirmed the superiority of the dual conjugate compared to the single HA-drug conjugates in suppressing the tumor growth. Immunoprofiling of the excised tumors revealed a significant increase in the M1/M2 ratio of TAMs in mice treated with the dual conjugate. Our intravenously injectable HA conjugate of RES and BEX provides a promising immunotherapeutic combination strategy for resetting the M1/M2 ratio, supporting the tumoricidal activity of TAMs for effective melanoma treatment.
Collapse
Affiliation(s)
- Marwa A Sallam
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Industrial pharmacy, Faculty of pharmacy, Alexandria University, 21521, Egypt
| | - C Wyatt Shields Iv
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Supriya Prakash
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Jayoung Kim
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Daniel C Pan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
10
|
Raviraj V, Pham BTT, Kim BJ, Pham NTH, Kok LF, Painter N, Delic NC, Jones SK, Hawkett BS, Lyons JG. Non-invasive transdermal delivery of chemotherapeutic molecules in vivo using superparamagnetic iron oxide nanoparticles. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00079-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
The skin is both a target and a potential conduit for the delivery of drugs, but its cornified cell layer resists penetration by most molecules. This study investigated the potential of superparamagnetic iron oxide nanoparticles to facilitate the transdermal delivery of anticancer agents.
Results
Chemotherapeutic cancer drugs were applied with or without nanoparticles to the skin of hairless mice, and their ability to penetrate the skin was assessed using fluorescence microscopy and tumor growth. Nanoparticles enhanced the penetration of the skin by doxorubicin and 5-fluorouracil as determined by fluorescence microscopy and growth retardation of experimental melanoma in immunocompetent, syngeneic mice. This drug enhancement did not require conjugation or encapsulation of the drugs by the nanoparticles—simple co-administration sufficed. Nanoparticles applied topically to melanomas increased the cytotoxicity and immune cell infiltration induced by co-administered 5-fluorouracil, and also reduced vascularization of the tumors independently of 5-fluorouracil.
Conclusion
Correctly formulated superparamagnetic iron oxide nanoparticles can facilitate the chemotherapeutic effectiveness of cytotoxic drugs on skin tumors by both increasing their transdermal penetration and ameliorating host–tumor interactions. This enhancement of skin penetration occurs without the need for conjugation or encapsulation of the co-administered drugs, and so will likely be applicable to other drugs, also.
Collapse
|
11
|
Fakhri KU, Sultan A, Mushtaque M, Hasan MR, Nafees S, Hafeez ZB, Zafaryab M, Rizwanullah M, Sharma D, Bano F, AlMalki WH, Ahmad FJ, Rizvi MMA. Obstructions in Nanoparticles Conveyance, Nano-Drug Retention, and EPR Effect in Cancer Therapies. HANDBOOK OF RESEARCH ON ADVANCEMENTS IN CANCER THERAPEUTICS 2021. [DOI: 10.4018/978-1-7998-6530-8.ch026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this chapter, the authors first review nano-devices that are mixtures of biologic molecules and synthetic polymers like nano-shells and nano-particles for the most encouraging applications for different cancer therapies. Nano-sized medications additionally spill especially into tumor tissue through penetrable tumor vessels and are then held in the tumor bed because of diminished lymphatic drainage. This procedure is known as the enhanced penetrability and retention (EPR) impact. Nonetheless, while the EPR impact is generally held to improve conveyance of nano-medications to tumors, it in certainty offers not exactly a 2-overlay increment in nano-drug conveyance contrasted with basic ordinary organs, bringing about medication concentration that is not adequate for restoring most malignant growths. In this chapter, the authors likewise review different obstructions for nano-sized medication conveyance and to make the conveyance of nano-sized medications to tumors progressively successful by expanding on the EPR impact..
Collapse
Affiliation(s)
| | | | | | | | | | | | - Md Zafaryab
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Md Rizwanullah
- School of Pharmaceutical Education and Research, Jamia Hamdard, India
| | - Deepti Sharma
- Institute of Nuclear Medicine and Allied Sciences, India
| | - Farhad Bano
- National Institute of Immunology, New Delhi, India
| | | | - Farhan Jalees Ahmad
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
12
|
Liu YR, Cheng YQ, Wang SB, Su YR, Liu Y, Li CY, Jin L, Wan Q, Sang X, Wang ZC. Therapeutic effects and perspective of stem cell extracellular vesicles in aging and cancer. J Cell Physiol 2020; 236:4783-4796. [PMID: 33368322 DOI: 10.1002/jcp.30212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/02/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
Senescent cells can secrete a plethora of cytokines which induce senescent phenotype of neighboring cells and was called senescence-associated secretory phenotype. Previously, it was believed that cancer was caused by the infinite division and uncontrolled proliferation of cells. Based on this, anticancer treatments were all aimed at killing cancer cells. Cancer is now considered an age-related disease. Cancer cells are not exogenous, but one of the worst results of injuries which initially induce cell senescence. Therefore, reversing cell senescence can fundamentally prevent and treat cancer. Though current anticancer treatments induce the cancer cells apoptosis, they induce senescence of normal cells at the same time, thus promoting the occurrence and development of cancer and forming a vicious circle. Extracellular vesicles (EVs) are nano-sized vesicles which partially mirror their parent cells. In the tumor microenvironment, EVs of senescent cells can change the expression profile of cancer cells, contributing to their resistance to chemotherapy. There is growing evidence indicates that stem cell EVs exert effective antiaging and anticancer actions by transferring functional microRNAs and proteins. This review will summarize the therapeutic role of stem cell EVs in reversing aging and cancer, which suggests the broad clinical application perspective.
Collapse
Affiliation(s)
- Yu-Run Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ya-Qi Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shou-Bi Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ya-Ru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Chao-Yang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qi Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xuan Sang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Chong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Sebak AA, Gomaa IEO, ElMeshad AN, Farag MH, Breitinger U, Breitinger HG, AbdelKader MH. Distinct Proteins in Protein Corona of Nanoparticles Represent a Promising Venue for Endogenous Targeting - Part II: In vitro and in vivo Kinetics Study. Int J Nanomedicine 2020; 15:9539-9556. [PMID: 33299308 PMCID: PMC7721286 DOI: 10.2147/ijn.s273721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/30/2020] [Indexed: 01/04/2023] Open
Abstract
Introduction Nanoparticles (NPs), upon introduction to the biological systems, become wrapped by serum and cellular proteins constituting the protein corona (PC). This PC contributes largely to the NPs’ interaction with the biological systems and their subsequent functions. On the one hand, PC can decrease the efficiency of targeting by directing the NPs to the reticuloendothelial system (RES) or by masking the active targeting moieties and decreasing their ability to bind to their target receptors. On the other hand, some components of PC have offered hopes for achieving endogenous targeting. Methods In this study, we aimed at the investigation of the role of the PC in determining the behavior of cRGDyk peptide-unconjugated and -conjugated NPs (uNPs and cNPs) exhibiting different physicochemical properties and their interaction with melanoma on in vitro and in vivo levels. Mathematical modeling has been utilized to understand the kinetics of the interaction of NPs with the tumor cells and different organs, respectively. Results Endocytosis and exocytosis were reported to occur simultaneously for the utilized NPs. The balance was largely dependent on the NPs’ physicochemical properties and the role of the PC. In addition, distinct proteins present in the PC (illustrated in the results of the PC analysis in part I) have also determined the patterns of the NPs’ distribution in different organs and tissues of the vascular system, the RES system and the target tumot tissue. Vitronectin (VN) was found to mediate higher accumulation in integrin receptor-expressing melanoma cells, while complement 3 protein (C3) and clusterin (CLU), as an opsonin and dysopsonin, respectively, regulated the balance between the RES uptake and blood circulation. Discussion PC, if properly modulated by tuning NPs’ physicochemical properties, can serve as a potential venue for optimum utilization of NPs in cancer therapy.
Collapse
Affiliation(s)
- Aya Ahmed Sebak
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Iman Emam Omar Gomaa
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Aliaa Nabil ElMeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mahmoud Hussien Farag
- Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Ulrike Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Hans-Georg Breitinger
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo City, Egypt
| | - Mahmoud Hashem AbdelKader
- National Institute of Laser Enhanced Sciences (NILES), Cairo University (CU), Giza, Egypt.,European University in Egypt (EUE), New Administrative Capital, Cairo, Egypt
| |
Collapse
|
14
|
Combination therapy with TiO 2 nanoparticles and cisplatin enhances chemotherapy response in murine melanoma models. Clin Transl Oncol 2020; 23:738-749. [PMID: 32734535 DOI: 10.1007/s12094-020-02463-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/17/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Despite recent progressions in the treatment of melanoma, the response to conventional therapies and the long-term survival in melanoma patients still remain poor. Recently, the use of nanoparticles (NPs) has been highlighted for promoting the chemotherapeutic effects of cytotoxic drugs in melanoma. The aim of this study is to mechanistically evaluate the potential of titanium dioxide (TiO2) nanoparticles (NPs) for enhancing chemotherapy effects in in vitro and in vivo models of murine melanoma. METHODS The F10 melanoma cells were exposed to different concentrations of TiO2 NPs and/or cisplatin, then cell growth, cell viability, and cell death were evaluated. In parallel, C57BL/6 syngeneic melanoma mice were treated by TiO2 NPs and/or cisplatin, and then drug responses, tumor size and mice's organs were studied pathologically. Autophagy was examined by evaluating the formation of autophagosomes and gene expression levels of autophagy markers (ATG5 and ATG6) by fluorescent microscopy and qPCR, respectively. RESULTS Nontoxic concentrations of TiO2 NPs (50 µg/ml) promote anti-proliferative and cytotoxic effects of cisplatin in F10 melanoma cells, which is mediated through the induction of autophagy and necrotic cell death. Whereas TiO2 NPs have no cytotoxic or metastatic effects in melanoma mice, its combination with cisplatin enhances drug responses (up to 50%), leading to higher inhibition of tumor growth compared with each monotherapy. CONCLUSION The combination of TiO2 NP with cisplatin enhances chemotherapy response in both in vitro and in vivo melanoma models. In addition, autophagy plays an essential role during sensitizing melanoma cells to chemotherapy.
Collapse
|
15
|
Chillà A, Margheri F, Biagioni A, Del Rosso T, Fibbi G, Del Rosso M, Laurenzana A. Cell-Mediated Release of Nanoparticles as a Preferential Option for Future Treatment of Melanoma. Cancers (Basel) 2020; 12:cancers12071771. [PMID: 32630815 PMCID: PMC7408438 DOI: 10.3390/cancers12071771] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 01/15/2023] Open
Abstract
Targeted and immune therapies have unquestionably improved the prognosis of melanoma patients. However the treatment of this neoplasm still requires approaches with a higher therapeutic index, in order to reduce shortcomings related to toxic effects and aspecific targeting. This means developing therapeutic tools derived with high affinity molecules for tumor components differentially expressed in melanoma cells with respect to their normal counterpart. Nanomedicine has sought to address this problem owing to the high modulability of nanoparticles. This approach exploits not only the enhanced permeability and retention effect typical of the tumor microenvironment (passive targeting), but also the use of specific "molecular antennas" that recognize some tumor-overexpressed molecules (active targeting). This line of research has given rise to the so-called "smart nanoparticles," some of which have already passed the preclinical phase and are under clinical trials in melanoma patients. To further improve nanoparticles partition within tumors, for some years now a line of thought is exploiting the molecular systems that regulate the innate tumor-homing activity of platelets, granulocytes, monocytes/macrophages, stem cells, endothelial-colony-forming cells, and red blood cells loaded with nanoparticles. This new vision springs from the results obtained with some of these cells in regenerative medicine, an approach called "cell therapy." This review takes into consideration the advantages of cell therapy as the only one capable of overcoming the limits of targeting imposed by the increased interstitial pressure of tumors.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Tommaso Del Rosso
- Department of Physics, Pontifical Catholic University of Rio de Janeiro, 22451-900 Rio de Janeiro-RJ, Brazil;
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
- Correspondence: (M.D.R.); (A.L.)
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
- Correspondence: (M.D.R.); (A.L.)
| |
Collapse
|
16
|
Abstract
Melanoma is a life-threatening disease caused by mutations in pigment-producing cells. Numerous treatments for melanoma have been approved in the past several decades; however, they often cause severe side effects and in most cases do not result in a complete cure. mRNA (messenger RNA) as a therapeutic agent provides a new avenue for melanoma treatment and several advantages over conventional treatments. The first mRNA drugs for melanoma treatment are currently in clinical trials, and approval of mRNA drugs by the Food and Drug Administration seems to be within reach. This new class of drugs can be readily adapted to other diseases, raising the hope of providing a new therapeutic option for various diseases.
Collapse
Affiliation(s)
- Melissa van Dülmen
- Department of Chemistry and Pharmacy, Institute for Biochemistry, University of Münster, Wilhelm-Klemm-Straße 2, 48149 Münster, Germany
| | - Andrea Rentmeister
- Department of Chemistry and Pharmacy, Institute for Biochemistry, University of Münster, Wilhelm-Klemm-Straße 2, 48149 Münster, Germany.,Cells in Motion Interfaculty Center, Waldeyerstraße 15, 48149 Münster, Germany
| |
Collapse
|
17
|
Zhu C, Zhu Y, Pan H, Chen Z, Zhu Q. Current Progresses of Functional Nanomaterials for Imaging Diagnosis and Treatment of Melanoma. Curr Top Med Chem 2019; 19:2494-2506. [PMID: 31642783 DOI: 10.2174/1568026619666191023130524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
Abstract
Melanoma is a malignant skin tumor that results in poor disease prognosis due to unsuccessful
treatment options. During the early stages of tumor progression, surgery is the primary approach
that assures a good outcome. However, in the presence of metastasis, melanoma hasbecome almost
immedicable, since the tumors can not be removed and the disease recurs easily in a short period of
time. However, in recent years, the combination of nanomedicine and chemotherapeutic drugs has offered
promising solutions to the treatment of late-stage melanoma. Extensive studies have demonstrated
that nanomaterials and their advanced applications can improve the efficacy of traditional chemotherapeutic
drugs in order to overcome the disadvantages, such as drug resistance, low drug delivery rate and
reduced targeting to the tumor tissue. In the present review, we summarized the latest progress in imaging
diagnosis and treatment of melanoma using functional nanomaterials, including polymers,
liposomes, metal nanoparticles, magnetic nanoparticles and carbon-based nanoparticles. These
nanoparticles are reported widely in melanoma chemotherapy, gene therapy, immunotherapy, photodynamic
therapy, and hyperthermia.
Collapse
Affiliation(s)
- Congcong Zhu
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Yunjie Zhu
- Cellular Biomedicine Group Inc., Shanghai 201210, China
| | - Huijun Pan
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Zhongjian Chen
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Quangang Zhu
- Department of Pharmacy, Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
18
|
Kohle FFE, Li S, Turker MZ, Wiesner UB. Ultrasmall PEGylated and Targeted Core-Shell Silica Nanoparticles Carrying Methylene Blue Photosensitizer. ACS Biomater Sci Eng 2019; 6:256-264. [PMID: 33463188 DOI: 10.1021/acsbiomaterials.9b01359] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Photodynamic therapy (PDT) presents an alternative noninvasive therapeutic modality for the treatment of cancer and other diseases. PDT relies on cytotoxic singlet oxygen (reactive oxygen species or ROS) that is locally generated through energy transfer between a photosensitizer (PS) and molecularly dissolved triplet oxygen. While a number of nanoparticle-based PS vehicles have been described, because of their beneficial and proven biodistribution and pharmacokinetic profiles, ultrasmall nanoparticles with diameters below 10 nm are particularly promising. Here, we investigate two different particle designs deviating from ultrasmall poly(ethylene glycol)-coated (PEGylated) fluorescent core-shell silica nanoparticles referred to as Cornell prime dots (C' dots) by replacing the fluorescent dye with a photosensitizer (psC' dots), here the methylene blue (MB) derivate MB2. In the first approach (design 1), MB2 is encapsulated into the matrix of the silica core, while in the second approach (design 2), MB2 is grafted onto the silica core surface in between chains of the sterically stabilizing poly(ethylene glycol) (PEG) corona. We compare both cases with regard to their singlet oxygen quantum yields, ΦΔ, with the effective ΦΔeff per particle reaching 111 ± 3 and 161 ± 5% for designs 1 and 2, respectively, substantially exceeding single MB2 molecule performance. Encapsulation significantly improves PS photostability, while surface conjugation diminishes it, relative to free MB2. Finally, we show that both particle designs allow functionalization with a targeting peptide, cyclo(Arg-Gly-Asp-D-Tyr-Cys) [c(RGDyC)]. Results suggest that psC' dots are a promising targeted platform for PDT applications, e.g. in oncology, that may combine colloidal stability, efficient renal clearance limiting off-target accumulation, targeted delivery to sites of disease, and effective ROS generation maximizing therapeutic efficacy.
Collapse
|
19
|
Shi X, Ma X, Ren E, Zhang Y, Jia D, Gao Y, Xue P, Kang Y, Liu G, Xu Z. Tumor-Microenvironment-Activatable Nanoreactor Based on a Polyprodrug for Multimodal-Imaging-Medicated Enhanced Cancer Chemo/Phototherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:40704-40715. [PMID: 31577408 DOI: 10.1021/acsami.9b16054] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Anticancer nanomedicine-based multimodal imaging and synergistic therapy hold great promise in cancer diagnosis and therapy owing to their abilities to improve therapeutic efficiency and reduce unnecessary side effects, producing promising clinical prospects. Herein, we integrated chemotherapeutic drug camptothecin (CPT) and near-infrared-absorbing new indocyanine green (IR820) into a single system by charge interaction and obtained a tumor-microenvironment-activatable PCPTSS/IR820 nanoreactor to perform thermal/fluorescence/photoacoustic-imaging-guided chemotherapy and photothermal therapy simultaneously. Specifically, the generated PCPTSS/IR820 showed an excellent therapeutic agent loading content and size stability, and the trials in vitro and in vivo suggested that the smart PCPTSS/IR820 could deeply permeate into tumor tissues due to its suitable micellar size. Upon near-infrared laser irradiation, the nanoreactor further produced a terrific synergism of chemo-photo treatment for cancer therapy. Therefore, the PCPTSS/IR820 polyprodrug-based nanoreactor holds outstanding promise for multimodal imaging and combined dual therapy.
Collapse
Affiliation(s)
| | | | - En Ren
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , P. R. China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , P. R. China
| | | | | | | | | | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health , Xiamen University , Xiamen 361102 , P. R. China
| | | |
Collapse
|
20
|
Abstract
Malignant melanoma is an aggressive skin cancer with limited therapeutic options. Cancer is the second largest cause of death in society and one of the most difficult diseases to treat. Advances in biotechnology have enabled the current use of nanotechnology via the application of nanomaterials, especially as drug delivery systems for the transportation of very small particles. In this context, carbon nanotubes, with a potential role in the diagnosis and treatment of melanoma, are still an emerging research field. Their properties have been extensively studied for the use of antineoplastics drugs, as well as for DNA and RNA interference for the treatment of cancer. However, the most important challenge in nanomedicine is to decrease the toxicity and increase the biocompatibility of the nanomaterials used to transport therapeutic molecules. In this sense, this article addresses the recent advances in the use of carbon nanotubes in melanoma therapy and highlights the opportunities and challenges in this area. The advances and challenges involving these topics are essential to the success of nanoconjugate systems, and studies improving the comprehension of these nanosystems contribute to the development of specific antitumor therapies.
Collapse
|
21
|
Feng X, Moy AJ, Nguyen HTM, Zhang Y, Zhang J, Fox MC, Sebastian KR, Reichenberg JS, Markey MK, Tunnell JW. Raman biophysical markers in skin cancer diagnosis. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-10. [PMID: 29752800 DOI: 10.1117/1.jbo.23.5.057002] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/23/2018] [Indexed: 05/22/2023]
Abstract
Raman spectroscopy (RS) has demonstrated great potential for in vivo cancer screening; however, the biophysical changes that occur for specific diagnoses remain unclear. We recently developed an inverse biophysical skin cancer model to address this issue. Here, we presented the first demonstration of in vivo melanoma and nonmelanoma skin cancer (NMSC) detection based on this model. We fit the model to our previous clinical dataset and extracted the concentration of eight Raman active components in 100 lesions in 65 patients diagnosed with malignant melanoma (MM), dysplastic nevi (DN), basal cell carcinoma, squamous cell carcinoma, and actinic keratosis. We then used logistic regression and leave-one-lesion-out cross validation to determine the diagnostically relevant model components. Our results showed that the biophysical model captures the diagnostic power of the previously used statistical classification model while also providing the skin's biophysical composition. In addition, collagen and triolein were the most relevant biomarkers to represent the spectral variances between MM and DN, and between NMSC and normal tissue. Our work demonstrates the ability of RS to reveal the biophysical basis for accurate diagnosis of different skin cancers, which may eventually lead to a reduction in the number of unnecessary excisional skin biopsies performed.
Collapse
Affiliation(s)
- Xu Feng
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, Unites States
| | - Austin J Moy
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, Unites States
| | - Hieu T M Nguyen
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, Unites States
| | - Yao Zhang
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, Unites States
| | - Jason Zhang
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, Unites States
| | - Matthew C Fox
- University of Texas at Austin, Dell Medical School, Department of Medicine, Austin, Texas, United States
| | - Katherine R Sebastian
- University of Texas at Austin, Dell Medical School, Department of Medicine, Austin, Texas, United States
| | - Jason S Reichenberg
- University of Texas at Austin, Dell Medical School, Department of Medicine, Austin, Texas, United States
| | - Mia K Markey
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, Unites States
| | - James W Tunnell
- University of Texas at Austin, Department of Biomedical Engineering, Austin, Texas, Unites States
| |
Collapse
|
22
|
Liu Q, Das M, Liu Y, Huang L. Targeted drug delivery to melanoma. Adv Drug Deliv Rev 2018; 127:208-221. [PMID: 28939379 DOI: 10.1016/j.addr.2017.09.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/29/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022]
Abstract
Melanoma derived from melanocytes is the most aggressive genre of skin cancer. Although the considerable advancement in the study of human cancer biology and drug discovery, most advanced melanoma patients are inevitably unable to be cured. With the emergence of nanotechnology, the use of nano-carriers is widely expected to alter the landscape of melanoma treatment. In this review, we will discuss melanoma biology, current treatment options, mechanisms behind drug resistance, and nano-based solutions for effective anti-cancer therapy, followed by challenges and perspectives in both pre-clinical and clinical settings.
Collapse
Affiliation(s)
- Qi Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC & NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
23
|
Naidoo C, Kruger CA, Abrahamse H. Photodynamic Therapy for Metastatic Melanoma Treatment: A Review. Technol Cancer Res Treat 2018; 17:1533033818791795. [PMID: 30099929 PMCID: PMC6090489 DOI: 10.1177/1533033818791795] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 06/04/2018] [Accepted: 07/03/2018] [Indexed: 01/21/2023] Open
Abstract
This review article is based on specifically targeted nanoparticles that have been used in the treatment of melanoma. According to the Skin Cancer Foundation, within 2017 an estimated 9730 people will die due to invasive melanoma. Conventional treatments for nonmalignant melanoma include surgery, chemotherapy, and radiation. For the treatment of metastatic melanoma, 3 therapeutic agents have been approved by the Food and Drug Administration: dacarbazine, recombinant interferon α-2b, and high-dose interleukin 2. Photodynamic therapy is an alternative therapy that activates a photosensitizer at a specific wavelength forming reactive oxygen species which in turn induces cell death; it is noninvasive with far less side effects when compared to conventional treatments. Nanoparticles are generally conjugated to photosynthetic drugs, since they are biocompatible, stabile, and durable, as well as have a high loading capacity, which improve either passive or active photosensitizer drug delivery to targeted cells. Therefore, various photosynthetic drugs and nanoparticle drug delivery systems specifically targeted for melanoma were analyzed in this review article in relation to either their passive or their active cellular uptake mechanisms in order to deduce the efficacy of photodynamic therapy treatment for metastatic melanoma which currently remains ongoing. The overall findings from this review concluded that no current photodynamic therapy studies have been performed in relation to active nanoparticle platform photosensitizer drug carrier systems for the treatment of metastatic melanoma, and so this type of research requires further investigation into developing a more efficient active nano-photosensitizer carrier smart drug that can be conjugated to specific cell surface receptors and combinative monoclonal antibodies so that a further enhanced and more efficient form of targeted photodynamic therapy for the treatment of metastatic melanoma can be established.
Collapse
Affiliation(s)
- Channay Naidoo
- Laser Research Centre, Faculty of Health Sciences, University of
Johannesburg, Johannesburg, South Africa
| | - Cherie Ann Kruger
- Laser Research Centre, Faculty of Health Sciences, University of
Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of
Johannesburg, Johannesburg, South Africa
| |
Collapse
|
24
|
Zhang C, Zhang J, Qin Y, Song H, Huang P, Wang W, Wang C, Li C, Wang Y, Kong D. Co-delivery of doxorubicin and pheophorbide A by pluronic F127 micelles for chemo-photodynamic combination therapy of melanoma. J Mater Chem B 2018; 6:3305-3314. [DOI: 10.1039/c7tb03179c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this work, doxorubicin (DOX)-loaded pheophorbide A (PheoA) modified Pluronic F127 (F127) micelles (DOX/F127-PheoA micelles) were developed for combined chemo-photodynamic therapy of melanoma.
Collapse
|
25
|
Li L, Pang X, Liu G. Near-Infrared Light-Triggered Polymeric Nanomicelles for Cancer Therapy and Imaging. ACS Biomater Sci Eng 2017; 4:1928-1941. [DOI: 10.1021/acsbiomaterials.7b00648] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Lei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xin Pang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
26
|
Rosa TG, dos Santos SN, de Jesus Andreoli Pinto T, Ghisleni DDM, Barja-Fidalgo TC, Ricci-Junior E, Al-Qahtani M, Kozempel J, Bernardes ES, Santos-Oliveira R. Microradiopharmaceutical for Metastatic Melanoma. Pharm Res 2017; 34:2922-2930. [DOI: 10.1007/s11095-017-2275-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 10/01/2017] [Indexed: 12/12/2022]
|
27
|
Naserzadeh P, Ansari Esfeh F, Kaviani M, Ashtari K, Kheirbakhsh R, Salimi A, Pourahmad J. Single-walled carbon nanotube, multi-walled carbon nanotube and Fe 2O 3 nanoparticles induced mitochondria mediated apoptosis in melanoma cells. Cutan Ocul Toxicol 2017; 37:157-166. [PMID: 28768445 DOI: 10.1080/15569527.2017.1363227] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Nanomaterials (NM) exhibit novel anticancer properties. MATERIALS AND METHODS The toxicity of three nanoparticles that are currently being produced in high tonnage including single-walled carbon nanotube (SWCNT), multi-walled carbon nanotube (MWCNT) and Fe2O3 nanoparticles, were compared with normal and melanoma cells. RESULTS All tested nanoparticles induced selective toxicity and caspase 3 activation through mitochondria pathway in melanoma cells and mitochondria cause the generating of reactive oxygen species (ROS), mitochondrial membrane potential decline (MMP collapse), mitochondria swelling, and cytochrome c release. The pretreatment of butylated hydroxytoluene (BHT), a cell-permeable antioxidant and cyclosporine A (Cs. A), a mitochondrial permeability transition (MPT), pore sealing agent decreased cytotoxicity, caspase 3 activation, ROS generation, and mitochondrial damages induced by SWCNT, MWCNT, and IONPs. CONCLUSIONS Our promising results provide a potential approach for the future therapeutic use of SWCNT, MWCNT, and IONPs in melanoma through mitochondrial targeting.
Collapse
Affiliation(s)
- Parvaneh Naserzadeh
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Fatemeh Ansari Esfeh
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mahboubeh Kaviani
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Khadijeh Ashtari
- d Department of Medical Nanotechnology, Faculty of Advanced Technology in Medicine , Iran University of Medical Sciences , Tehran , Iran
| | - Raheleh Kheirbakhsh
- b Cancer Biology Research Center , Cancer Institute of Iran, Tehran University of Medical Sciences , Tehran , Iran
| | - Ahmad Salimi
- c Department of Pharmacology and Toxicology, School of Pharmacy , Ardabil University of Medical Science , Ardabil , Iran
| | - Jalal Pourahmad
- a Department of Pharmacology and Toxicology, Faculty of Pharmacy , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
28
|
Badea I. New strategies in melanoma therapy: can nanoparticles overcome chemoresistance? Nanomedicine (Lond) 2017. [DOI: 10.2217/nnm-2017-0145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Ildiko Badea
- College of Pharmacy & Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Room 3D01.5, Box 3D01–13, Saskatoon, Saskatchewan, S7N 5E5, Canada
| |
Collapse
|
29
|
Zhang T, Prasad P, Cai P, He C, Shan D, Rauth AM, Wu XY. Dual-targeted hybrid nanoparticles of synergistic drugs for treating lung metastases of triple negative breast cancer in mice. Acta Pharmacol Sin 2017; 38:835-847. [PMID: 28216624 PMCID: PMC5520182 DOI: 10.1038/aps.2016.166] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/26/2016] [Indexed: 12/21/2022] Open
Abstract
Lung metastasis is the major cause of death in patients with triple negative breast
cancer (TNBC), an aggressive subtype of breast cancer with no effective therapy at
present. It has been proposed that dual-targeted therapy, ie, targeting
chemotherapeutic agents to both tumor vasculature and cancer cells, may offer some
advantages. The present work was aimed to develop a dual-targeted synergistic drug
combination nanomedicine for the treatment of lung metastases of TNBC. Thus,
Arg-Gly-Asp peptide (RGD)-conjugated, doxorubicin (DOX) and mitomycin C (MMC)
co-loaded polymer-lipid hybrid nanoparticles (RGD-DMPLN) were prepared and
characterized. The synergism between DOX and MMC and the effect of RGD-DMPLN on cell
morphology and cell viability were evaluated in human MDA-MB-231 cells in
vitro. The optimal RGD density on nanoparticles (NPs) was identified based on
the biodistribution and tumor accumulation of the NPs in a murine lung metastatic
model of MDA-MB-231 cells. The microscopic distribution of RGD-conjugated NPs in lung
metastases was examined using confocal microscopy. The anticancer efficacy of
RGD-DMPLN was investigated in the lung metastatic model. A synergistic ratio of DOX
and MMC was found in the MDA-MB-231 human TNBC cells. RGD-DMPLN induced morphological
changes and enhanced cytotoxicity in vitro. NPs with a median RGD density
showed the highest accumulation in lung metastases by targeting both tumor
vasculature and cancer cells. Compared to free drugs, RGD-DMPLN exhibited
significantly low toxicity to the host, liver and heart. Compared to non-targeted
DMPLN or free drugs, administration of RGD-DMPLN (10 mg/kg, iv) resulted in a
4.7-fold and 31-fold reduction in the burden of lung metastases measured by
bioluminescence imaging, a 2.4-fold and 4.0-fold reduction in the lung metastasis
area index, and a 35% and 57% longer median survival time, respectively.
Dual-targeted RGD-DMPLN, with optimal RGD density, significantly inhibited the
progression of lung metastasis and extended host survival.
Collapse
|
30
|
Tapia-Moreno A, Juarez-Moreno K, Gonzalez-Davis O, Cadena-Nava RD, Vazquez-Duhalt R. Biocatalytic virus capsid as nanovehicle for enzymatic activation of Tamoxifen in tumor cells. Biotechnol J 2017; 12. [DOI: 10.1002/biot.201600706] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/28/2017] [Accepted: 03/31/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Alejandro Tapia-Moreno
- Centro de Nanociencias y Nanotecnología; Universidad Nacional Autónoma de México; Ensenada Baja California Mexico
| | - Karla Juarez-Moreno
- Centro de Nanociencias y Nanotecnología; Universidad Nacional Autónoma de México; Ensenada Baja California Mexico
| | - Oscar Gonzalez-Davis
- Centro de Nanociencias y Nanotecnología; Universidad Nacional Autónoma de México; Ensenada Baja California Mexico
| | - Ruben D. Cadena-Nava
- Centro de Nanociencias y Nanotecnología; Universidad Nacional Autónoma de México; Ensenada Baja California Mexico
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología; Universidad Nacional Autónoma de México; Ensenada Baja California Mexico
| |
Collapse
|
31
|
Oh Y, Jin JO, Oh J. Photothermal-triggered control of sub-cellular drug accumulation using doxorubicin-loaded single-walled carbon nanotubes for the effective killing of human breast cancer cells. NANOTECHNOLOGY 2017; 28:125101. [PMID: 28145889 DOI: 10.1088/1361-6528/aa5d7d] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Single-walled carbon nanotubes (SWNTs) are often the subject of investigation as effective photothermal therapy (PTT) agents owing to their unique strong optical absorption. Doxorubicin (DOX)-loaded SWNTs (SWNTs-DOX) can be used as an efficient therapeutic agent for combined near infrared (NIR) cancer photothermal and chemotherapy. However, SWNTs-DOX-mediated induction of cancer cell death has not been fully investigated, particularly the reaction of DOX inside cancer cells by PTT. In this study, we examined how the SWNTs-DOX promoted effective MDA-MB-231 cell death compared to DOX and PTT alone. We successfully synthesized the SWNTs-DOX. The SWNTs-DOX exhibited a slow DOX release, which was accelerated by NIR irradiation. Furthermore, DOX released from the SWNTs-DOX accumulated inside the cells at high concentration and effectively localized into the MDA-MB-231 cell nucleus. A combination of SWNTs-DOX and PTT promoted an effective MDA-MB-231 cell death by mitochondrial disruption and ROS generation. Thus, SWNTs-DOX can be utilized as an excellent anticancer agent for early breast cancer treatment.
Collapse
Affiliation(s)
- Yunok Oh
- Department of Marine-Bio Convergence Science, Pukyong National University, Busan 48513, Republic of Korea
| | | | | |
Collapse
|
32
|
Deng C, Zhang Q, Fu Y, Sun X, Gong T, Zhang Z. Coadministration of Oligomeric Hyaluronic Acid-Modified Liposomes with Tumor-Penetrating Peptide-iRGD Enhances the Antitumor Efficacy of Doxorubicin against Melanoma. ACS APPLIED MATERIALS & INTERFACES 2017; 9:1280-1292. [PMID: 28009503 DOI: 10.1021/acsami.6b13738] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A safe and efficient tumor-targeting strategy based on oligomeric hyaluronic acid (HA) modification and coadministration of tumor-penetrating peptide-iRGD was successfully developed. In this study, common liposomes (cLip) were modified by oligomeric HA to obtain HA-Lip. After injection into rats, HA-Lip showed good stealth in the bloodstream and lower liver distribution compared with cLip. Moreover, our HA-Lip could be internalized into B16F10 cells (CD44-overexpressing tumor cells) through HA-CD44 interaction. After systemic administration to B16F10 melanoma-bearing mice, HA-Lip showed an increased distribution in tumor due to the prolonged blood circulation time and the enhanced penetration and retention effect. When coadministered with iRGD, the tumor penetration of HA-Lip was significantly enhanced, which could promote HA-Lip internalization by tumors cells located in deep tumor regions through receptor-mediated endocytosis. Furthermore, doxorubicin (DOX)-loaded HA-Lip coadministering with iRGD showed much better antitumor effect compared to DOX-loaded cLip and DOX-loaded cLip in combination with iRGD. In systemic toxicity test, DOX-loaded HA-Lip could significantly decrease the cardiotoxicity and myelosuppression of DOX. Taken together, our results demonstrated that coadministration of oligomeric HA-modified liposomes with iRGD could be a promising treatment strategy for targeted therapy of melanoma.
Collapse
Affiliation(s)
- Caifeng Deng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Quan Zhang
- School of Pharmacy, Chengdu Medical College , Chengdu 610083, China
| | - Yao Fu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Tao Gong
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University , Chengdu 610041, China
| |
Collapse
|
33
|
Application of aluminum chloride phthalocyanine-loaded solid lipid nanoparticles for photodynamic inactivation of melanoma cells. Int J Pharm 2017; 518:228-241. [PMID: 28063902 DOI: 10.1016/j.ijpharm.2017.01.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/16/2016] [Accepted: 01/02/2017] [Indexed: 12/12/2022]
Abstract
Cutaneous melanoma is the most aggressive skin cancer and is particularly resistant to current therapeutic approaches. Photodynamic therapy (PDT) is a well-established photoprocess that is employed to treat some cancers, including non-melanoma skin cancer. Aluminum chloride phthalocyanine (ClAlPc) is used as a photosensitizer in PDT; however, its high hydrophobicity hampers its photodynamic activity under physiological conditions. The aim of this study was to produce solid lipid nanoparticles (SLN) containing ClAlPc using the direct emulsification method. ClAlPc-loaded SLNs (ClAlPc/SLNs) were characterized according to their particle size and distribution, zeta potential, morphology, encapsulation efficiency, stability, and phototoxic action in vitro in B16-F10 melanoma cells. ClAlPc/SLN had a mean diameter between 100 and 200nm, homogeneous size distribution (polydispersity index <0.3), negative zeta potential, and spherical morphology. The encapsulation efficiency was approximately 100%. The lipid crystallinity was investigated using X-ray diffraction and differential scanning calorimetry and indicated that ClAlPc was integrated into the SLN matrix. The ClAlPc/SLN formulations maintained their physicochemical stability without expelling the drug over a 24-month period. Compared to free ClAlPc, ClAlPc/SLN exerted outstanding phototoxicity effects in vitro against melanoma cells. Therefore, our results demonstrated that the ClAlPc/SLN described in the current study has the potential for use in further preclinical and clinical trials in PDT for melanoma treatment.
Collapse
|
34
|
Theranostic barcoded nanoparticles for personalized cancer medicine. Nat Commun 2016; 7:13325. [PMID: 27830705 PMCID: PMC5109543 DOI: 10.1038/ncomms13325] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 09/21/2016] [Indexed: 02/08/2023] Open
Abstract
Personalized medicine promises to revolutionize cancer therapy by matching the most effective treatment to the individual patient. Using a nanoparticle-based system, we predict the therapeutic potency of anticancer medicines in a personalized manner. We carry out the diagnostic stage through a multidrug screen performed inside the tumour, extracting drug activity information with single cell sensitivity. By using 100 nm liposomes, loaded with various cancer drugs and corresponding synthetic DNA barcodes, we find a correlation between the cell viability and the drug it was exposed to, according to the matching barcodes. Based on this screen, we devise a treatment protocol for mice bearing triple-negative breast-cancer tumours, and its results confirm the diagnostic prediction. We show that the use of nanotechnology in cancer care is effective for generating personalized treatment protocols. Determining the most effective treatment for each cancer patient is a key challenge in cancer therapy. In this article, the authors show, in a mouse model of breast cancer, that DNA barcoded nanoparticles can be used for pre-screening the efficacy of anticancer drugs.
Collapse
|
35
|
Emerging concepts of T cell metabolism as a target of immunotherapy. Nat Immunol 2016; 17:364-8. [PMID: 27002844 DOI: 10.1038/ni.3415] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/17/2016] [Indexed: 12/27/2022]
Abstract
T cells have a pivotal protective role in defense against infection and cancer but also are instrumental in the development of many autoimmune diseases. The regulation of nutrient uptake and utilization in T cells is critically important for the control of their differentiation, and manipulating metabolic pathways in these cells can alter their function and longevity. While the importance of T cell metabolic remodeling in different physiological settings is not fully understood, there is a growing realization that inappropriate metabolic remodeling underlies many aberrant immune responses and that manipulating cellular metabolism can beneficially enhance or temper immunity. Here we comment on the basic metabolic pathways in T cells, followed by a discussion on up-to-date findings about the relationship between metabolism and T cell function and longevity. Furthermore, we expand on potential approaches and applications in which T cells might be manipulated by the reprogramming of metabolic pathways for therapeutic purposes.
Collapse
|
36
|
Wang H, Yu J, Lu X, He X. Nanoparticle systems reduce systemic toxicity in cancer treatment. Nanomedicine (Lond) 2015; 11:103-6. [PMID: 26653177 DOI: 10.2217/nnm.15.166] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Hai Wang
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA.,Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jianhua Yu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.,Division of Hematology, Department of Internal medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Xiongbin Lu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA.,Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|