1
|
Haque M, Shakil MS, Mahmud KM. The Promise of Nanoparticles-Based Radiotherapy in Cancer Treatment. Cancers (Basel) 2023; 15:cancers15061892. [PMID: 36980778 PMCID: PMC10047050 DOI: 10.3390/cancers15061892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Radiation has been utilized for a long time for the treatment of cancer patients. However, radiotherapy (RT) has many constraints, among which non-selectivity is the primary one. The implementation of nanoparticles (NPs) with RT not only localizes radiation in targeted tissue but also provides significant tumoricidal effect(s) compared to radiation alone. NPs can be functionalized with both biomolecules and therapeutic agents, and their combination significantly reduces the side effects of RT. NP-based RT destroys cancer cells through multiple mechanisms, including ROS generation, which in turn damages DNA and other cellular organelles, inhibiting of the DNA double-strand damage-repair system, obstructing of the cell cycle, regulating of the tumor microenvironment, and killing of cancer stem cells. Furthermore, such combined treatments overcome radioresistance and drug resistance to chemotherapy. Additionally, NP-based RT in combined treatments have shown synergistic therapeutic benefit(s) and enhanced the therapeutic window. Furthermore, a combination of phototherapy, i.e., photodynamic therapy and photothermal therapy with NP-based RT, not only reduces phototoxicity but also offers excellent therapeutic benefits. Moreover, using NPs with RT has shown promise in cancer treatment and shown excellent therapeutic outcomes in clinical trials. Therefore, extensive research in this field will pave the way toward improved RT in cancer treatment.
Collapse
Affiliation(s)
- Munima Haque
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Md Salman Shakil
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka 1212, Bangladesh
| | - Kazi Mustafa Mahmud
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| |
Collapse
|
2
|
Marcu LG, Moghaddasi L, Bezak E. Cannot Target What Cannot Be Seen: Molecular Imaging of Cancer Stem Cells. Int J Mol Sci 2023; 24:ijms24021524. [PMID: 36675033 PMCID: PMC9864237 DOI: 10.3390/ijms24021524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/29/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Cancer stem cells are known to play a key role in tumour development, proliferation, and metastases. Their unique properties confer resistance to therapy, often leading to treatment failure. It is believed that research into the identification, targeting, and eradication of these cells can revolutionise oncological treatment. Based on the principle that what cannot be seen, cannot be targeted, a primary step in cancer management is the identification of these cells. The current review aims to encompass the state-of-the-art functional imaging techniques that enable the identification of cancer stem cells via various pathways and mechanisms. The paper presents in vivo molecular techniques that are currently available or await clinical implementation. Challenges and future prospects are highlighted to open new research avenues in cancer stem cell imaging.
Collapse
Affiliation(s)
- Loredana G. Marcu
- Faculty of Informatics and Science, University of Oradea, 1 Universitatii Str., 410087 Oradea, Romania
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- Correspondence:
| | - Leyla Moghaddasi
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Eva Bezak
- Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
3
|
Mondal J, An JM, Surwase SS, Chakraborty K, Sutradhar SC, Hwang J, Lee J, Lee YK. Carbon Nanotube and Its Derived Nanomaterials Based High Performance Biosensing Platform. BIOSENSORS 2022; 12:731. [PMID: 36140116 PMCID: PMC9496036 DOI: 10.3390/bios12090731] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/30/2022]
Abstract
After the COVID-19 pandemic, the development of an accurate diagnosis and monitoring of diseases became a more important issue. In order to fabricate high-performance and sensitive biosensors, many researchers and scientists have used many kinds of nanomaterials such as metal nanoparticles (NPs), metal oxide NPs, quantum dots (QDs), and carbon nanomaterials including graphene and carbon nanotubes (CNTs). Among them, CNTs have been considered important biosensing channel candidates due to their excellent physical properties such as high electrical conductivity, strong mechanical properties, plasmonic properties, and so on. Thus, in this review, CNT-based biosensing systems are introduced and various sensing approaches such as electrochemical, optical, and electrical methods are reported. Moreover, such biosensing platforms showed excellent sensitivity and high selectivity against not only viruses but also virus DNA structures. So, based on the amazing potential of CNTs-based biosensing systems, healthcare and public health can be significantly improved.
Collapse
Affiliation(s)
- Jagannath Mondal
- Department of Green Bio Engineering, Korea National University of Transportation, Chungju 27469, Korea
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea
| | - Sachin S. Surwase
- 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Korea
| | - Kushal Chakraborty
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Korea
| | - Sabuj Chandra Sutradhar
- 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Korea
| | - Joon Hwang
- 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Korea
- Department of Aeronautical & Mechanical Design Engineering, Korea National University of Transportation, Chungju 27469, Korea
| | - Jaewook Lee
- 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Korea
| | - Yong-Kyu Lee
- Department of Green Bio Engineering, Korea National University of Transportation, Chungju 27469, Korea
- 4D Convergence Technology Institute, Korea National University of Transportation, Jungpyeong 27909, Korea
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea
| |
Collapse
|
4
|
Carbon nanotube as an emerging theranostic tool for oncology. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
5
|
Peserico A, Di Berardino C, Russo V, Capacchietti G, Di Giacinto O, Canciello A, Camerano Spelta Rapini C, Barboni B. Nanotechnology-Assisted Cell Tracking. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:1414. [PMID: 35564123 PMCID: PMC9103829 DOI: 10.3390/nano12091414] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023]
Abstract
The usefulness of nanoparticles (NPs) in the diagnostic and/or therapeutic sector is derived from their aptitude for navigating intra- and extracellular barriers successfully and to be spatiotemporally targeted. In this context, the optimization of NP delivery platforms is technologically related to the exploitation of the mechanisms involved in the NP-cell interaction. This review provides a detailed overview of the available technologies focusing on cell-NP interaction/detection by describing their applications in the fields of cancer and regenerative medicine. Specifically, a literature survey has been performed to analyze the key nanocarrier-impacting elements, such as NP typology and functionalization, the ability to tune cell interaction mechanisms under in vitro and in vivo conditions by framing, and at the same time, the imaging devices supporting NP delivery assessment, and consideration of their specificity and sensitivity. Although the large amount of literature information on the designs and applications of cell membrane-coated NPs has reached the extent at which it could be considered a mature branch of nanomedicine ready to be translated to the clinic, the technology applied to the biomimetic functionalization strategy of the design of NPs for directing cell labelling and intracellular retention appears less advanced. These approaches, if properly scaled up, will present diverse biomedical applications and make a positive impact on human health.
Collapse
Affiliation(s)
- Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy; (C.D.B.); (V.R.); (G.C.); (O.D.G.); (A.C.); (C.C.S.R.); (B.B.)
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Bura C, Mocan T, Grapa C, Mocan L. Carbon Nanotubes-Based Assays for Cancer Detection and Screening. Pharmaceutics 2022; 14:pharmaceutics14040781. [PMID: 35456615 PMCID: PMC9028434 DOI: 10.3390/pharmaceutics14040781] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023] Open
Abstract
Carbon nanotubes (CNTs) were considered a potential cargo for cancer therapy and diagnosis following researchers’ shared goal of finding a new delivery system to enhance the pharmacological performance of the administered drugs. To date, several excellent reviews have focused on the role of CNTs as drug delivery systems, although there is currently no existing study that gathers all the advances in research-connected carbon nanotubes-based assay development for the early detection of cancer. In this review article, we will focus on the emerging role of CNTs as anticancer detection agents.
Collapse
Affiliation(s)
- Cristina Bura
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology ‘’Octavian Fodor’’, 400008 Cluj-Napoca, Romania; (C.B.); (T.M.); (C.G.)
| | - Teodora Mocan
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology ‘’Octavian Fodor’’, 400008 Cluj-Napoca, Romania; (C.B.); (T.M.); (C.G.)
- Department of Physiology, University of Medicine and Pharmacy, “Iuliu Hatieganu”, 400008 Cluj-Napoca, Romania
| | - Cristiana Grapa
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology ‘’Octavian Fodor’’, 400008 Cluj-Napoca, Romania; (C.B.); (T.M.); (C.G.)
- Department of Physiology, University of Medicine and Pharmacy, “Iuliu Hatieganu”, 400008 Cluj-Napoca, Romania
| | - Lucian Mocan
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology ‘’Octavian Fodor’’, 400008 Cluj-Napoca, Romania; (C.B.); (T.M.); (C.G.)
- Department of Surgery, University of Medicine and Pharmacy, “Iuliu Hatieganu”, 400008 Cluj-Napoca, Romania
- Correspondence:
| |
Collapse
|
7
|
Dou K, Feng W, Fan C, Cao Y, Xiang Y, Liu Z. Flexible Designing Strategy to Construct Activatable NIR-II Fluorescent Probes with Emission Maxima beyond 1200 nm. Anal Chem 2021; 93:4006-4014. [DOI: 10.1021/acs.analchem.0c04990] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Kun Dou
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Wenqi Feng
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Chen Fan
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Yu Cao
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules and College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Yunhui Xiang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Zhihong Liu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei 430072, China
- Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules and College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
8
|
Rai A, Noor S, Ahmad SI, Alajmi MF, Hussain A, Abbas H, Hasan GM. Recent Advances and Implication of Bioengineered Nanomaterials in Cancer Theranostics. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:91. [PMID: 33494239 PMCID: PMC7909769 DOI: 10.3390/medicina57020091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023]
Abstract
Cancer is one of the most common causes of death and affects millions of lives every year. In addition to non-infectious carcinogens, infectious agents contribute significantly to increased incidence of several cancers. Several therapeutic techniques have been used for the treatment of such cancers. Recently, nanotechnology has emerged to advance the diagnosis, imaging, and therapeutics of various cancer types. Nanomaterials have multiple advantages over other materials due to their small size and high surface area, which allow retention and controlled drug release to improve the anti-cancer property. Most cancer therapies have been known to damage healthy cells due to poor specificity, which can be avoided by using nanosized particles. Nanomaterials can be combined with various types of biomaterials to make it less toxic and improve its biocompatibility. Based on these properties, several nanomaterials have been developed which possess excellent anti-cancer efficacy potential and improved diagnosis. This review presents the latest update on novel nanomaterials used to improve the diagnostic and therapeutic of pathogen-associated and non-pathogenic cancers. We further highlighted mechanistic insights into their mode of action, improved features, and limitations.
Collapse
Affiliation(s)
- Ayushi Rai
- Department of Nanoscience, Central University of Gujarat, Sector 29, Gandhinagar 382030, India;
| | - Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India;
| | - Syed Ishraque Ahmad
- Department of Chemistry, Zakir Husain Delhi College, University of Delhi, New Delhi 110002, India;
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.)
| | - Hashim Abbas
- Department of Medicine, Nottingham University Hospitals, NHS Trust, Nottingham NG7 2UH, UK;
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
9
|
Kukkar D, Kukkar P, Kumar V, Hong J, Kim KH, Deep A. Recent advances in nanoscale materials for antibody-based cancer theranostics. Biosens Bioelectron 2020; 173:112787. [PMID: 33190049 DOI: 10.1016/j.bios.2020.112787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
The quest for advanced management tools or options of various cancers has been on the rise to efficiently reduce their risks of mortality without the demerits of conventional treatments (e.g., undesirable side effects of the medications on non-target tissues, non-targeted distribution, slow clearance of the administered drugs, and the development of drug resistance over the duration of therapy). In this context, nanomaterials-antibody conjugates can offer numerous advantages in the development of cancer theranostics over conventional delivery systems (e.g., highly specific and enhanced biodistribution of the drug in targeted tissues, prolonged systemic circulation, low toxicity, and minimally invasive molecular imaging). This review comprehensively discusses and evaluates recent advances in the application of nanomaterial-antibody bioconjugates for cancer theranostics for the further advancement in the control of diverse cancerous diseases. Further, discussion is expanded to cover the various challenges and limitations associated with the design and development of nanomaterial-antibody conjugates applicable towards better management of cancer.
Collapse
Affiliation(s)
- Deepak Kukkar
- Department of Nanotechnology, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, 140406, India
| | - Preeti Kukkar
- Department of Chemistry, Mata Gujri College, Fatehgarh Sahib, Punjab, 140406, India
| | - Vanish Kumar
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Punjab, 140306, India
| | - Jongki Hong
- College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Seoul, 02447, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, Seoul, 04763 Republic of Korea.
| | - Akash Deep
- Central Scientific Instruments Organization (CSIR-CSIO), Sector 30 C, Chandigarh, 160030, India.
| |
Collapse
|
10
|
Dzobo K, Senthebane DA, Ganz C, Thomford NE, Wonkam A, Dandara C. Advances in Therapeutic Targeting of Cancer Stem Cells within the Tumor Microenvironment: An Updated Review. Cells 2020; 9:E1896. [PMID: 32823711 PMCID: PMC7464860 DOI: 10.3390/cells9081896] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 12/24/2022] Open
Abstract
Despite great strides being achieved in improving cancer patients' outcomes through better therapies and combinatorial treatment, several hurdles still remain due to therapy resistance, cancer recurrence and metastasis. Drug resistance culminating in relapse continues to be associated with fatal disease. The cancer stem cell theory posits that tumors are driven by specialized cancer cells called cancer stem cells (CSCs). CSCs are a subpopulation of cancer cells known to be resistant to therapy and cause metastasis. Whilst the debate on whether CSCs are the origins of the primary tumor rages on, CSCs have been further characterized in many cancers with data illustrating that CSCs display great abilities to self-renew, resist therapies due to enhanced epithelial to mesenchymal (EMT) properties, enhanced expression of ATP-binding cassette (ABC) membrane transporters, activation of several survival signaling pathways and increased immune evasion as well as DNA repair mechanisms. CSCs also display great heterogeneity with the consequential lack of specific CSC markers presenting a great challenge to their targeting. In this updated review we revisit CSCs within the tumor microenvironment (TME) and present novel treatment strategies targeting CSCs. These promising strategies include targeting CSCs-specific properties using small molecule inhibitors, immunotherapy, microRNA mediated inhibitors, epigenetic methods as well as targeting CSC niche-microenvironmental factors and differentiation. Lastly, we present recent clinical trials undertaken to try to turn the tide against cancer by targeting CSC-associated drug resistance and metastasis.
Collapse
Affiliation(s)
- Kevin Dzobo
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Medical Campus, Anzio Road, Observatory, Cape Town 7925, South Africa; (D.A.S.); (C.G.)
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Dimakatso Alice Senthebane
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Medical Campus, Anzio Road, Observatory, Cape Town 7925, South Africa; (D.A.S.); (C.G.)
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Chelene Ganz
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Medical Campus, Anzio Road, Observatory, Cape Town 7925, South Africa; (D.A.S.); (C.G.)
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
| | - Nicholas Ekow Thomford
- Division of Human Genetics, Department of Pathology and Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa; (N.E.T.); (A.W.); (C.D.)
- Department of Medical Biochemistry, School of Medical Sciences, College of Health Sciences, University of Cape Coast, PMB, Cape Coast, Ghana
| | - Ambroise Wonkam
- Division of Human Genetics, Department of Pathology and Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa; (N.E.T.); (A.W.); (C.D.)
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology and Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa; (N.E.T.); (A.W.); (C.D.)
| |
Collapse
|
11
|
Jariyal H, Gupta C, Bhat VS, Wagh JR, Srivastava A. Advancements in Cancer Stem Cell Isolation and Characterization. Stem Cell Rev Rep 2020; 15:755-773. [PMID: 31863337 DOI: 10.1007/s12015-019-09912-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Occurrence of stem cells (CSCs) in cancer is well established in last two decades. These rare cells share several properties including presence of common surface markers, stem cell markers, chemo- and radio- resistance and are highly metastatic in nature; thus, considered as valuable prognostic and therapeutic targets in cancer. However, the studies related to CSCs pave number of issues due to rare cell population and difficulties in their isolation ascribed to common stem cell marker. Various techniques including flow cytometry, laser micro-dissection, fluorescent nanodiamonds and microfluidics are used for the isolation of these rare cells. In this review, we have included the advance strategies adopted for the isolation of CSCs using above mentioned techniques. Furthermore, CSCs are primarily found in the core of the solid tumors and their microenvironment plays an important role in maintenance, self-renewal, division and tumor development. Therefore, in vivo tracking and model development become obligatory for functional studies of CSCs. Fluorescence and bioluminescence tagging has been widely used for transplantation assay and lineage tracking experiments to improve our understanding towards CSCs behaviour in their niche. Techniques such as Magnetic resonance imaging (MRI) and Positron emission tomography (PET) have proved useful for tracking of endogenous CSCs which could be helpful in their identification in clinical settings.
Collapse
Affiliation(s)
- Heena Jariyal
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Chanchal Gupta
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Vedika Sandeep Bhat
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Jayant Ramakant Wagh
- Department of Biotechnology, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Akshay Srivastava
- Department of Medical Device, National institute of Pharmaceutical Education and Research -Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India.
| |
Collapse
|
12
|
Ghaffari H, Atashzar MR, Abdollahi H. Molecular imaging in tracking cancer stem cells: A review. Med J Islam Repub Iran 2020; 34:90. [PMID: 33306061 PMCID: PMC7711048 DOI: 10.34171/mjiri.34.90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Indexed: 11/05/2022] Open
Abstract
Cancer stem cells (CSCs) have critical roles in tumor development, progression, and recurrence. They are responsible for current cancer treatment failure and remain questionable for the design and development of new therapeutic strategies. With this issue, medical imaging provides several clues for finding biological mechanisms and strategies to treat CSCs. This review aims to summarize current molecular imaging approaches for detecting CSCs. In addition, some promising issues for CSCs finding and explaining biological mechanisms have been addressed. Among the molecular imaging approaches, modalities including Magnetic resonance imaging (MRI) and positron emission tomography (PET) have the greatest roles and several new approaches such as optical imaging are in progress.
Collapse
Affiliation(s)
- Hamed Ghaffari
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamid Abdollahi
- Department of Radiologic Sciences and Medical Physics, School of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
13
|
Liu J, Ma X, LingWang. Modulating effect of graphine oxide loaded hesperidin nanocomposite on the 1,2-dimethylhydrazine provoked colon carcinogenesis in rats via inhibiting the iNOS and COX-2 pathways. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2020.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
14
|
Sevic I, Spinelli FM, Vitale DL, Icardi A, Romano L, Brandone A, Giannoni P, Cristina C, Bolontrade MF, Alaniz L. Hyaluronan Metabolism is Associated with DNA Repair Genes in Breast and Colorectal Cancer. Screening of Potential Progression Markers Using qPCR. Biomedicines 2020; 8:E183. [PMID: 32610620 PMCID: PMC7400093 DOI: 10.3390/biomedicines8070183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022] Open
Abstract
In this work, we compared mRNA levels of Hyaluronan (HA) metabolism members and BRCA genes, known to be involved in the tumoral process, between tumor and non-tumor adjacent tissue and its correlation with previously proposed biomarkers (ER, PR, HER2 and KI67) in order to assess their value as a progression biomarkers. We show alteration in HA metabolism in colorectal but not breast cancer. However, we found a decrease in Hyaluronidase 1 HYAL1 levels in the breast but not colorectal cancer. We also show lower HA levels in tumor compared with normal tissue that could indicate a possible influence of tumor on its surrounding "normal" tissue. In both breast and colorectal cancer, CD44 and BRCA2 showed a strong positive correlation. Besides, our results show first indicators that qPCR of the analyzed genes could be used as an easy and low cost procedure for the evaluation of molecular markers we propose here.
Collapse
Affiliation(s)
- Ina Sevic
- Laboratorio de Microambiente Tumoral; Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín B6000, Argentina; (I.S.); (F.M.S.); (D.L.V.); (A.I.)
| | - Fiorella Mercedes Spinelli
- Laboratorio de Microambiente Tumoral; Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín B6000, Argentina; (I.S.); (F.M.S.); (D.L.V.); (A.I.)
| | - Daiana Lujan Vitale
- Laboratorio de Microambiente Tumoral; Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín B6000, Argentina; (I.S.); (F.M.S.); (D.L.V.); (A.I.)
| | - Antonella Icardi
- Laboratorio de Microambiente Tumoral; Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín B6000, Argentina; (I.S.); (F.M.S.); (D.L.V.); (A.I.)
| | - Lucia Romano
- Laboratorio de Fisiopatología de la Hipófisis; Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín B6000, Argentina; (L.R.); (C.C.)
| | - Alejandra Brandone
- Hospital Interzonal General de Agudos Dr. Abraham F. Piñeyro, Junín B6000, Argentina;
| | | | - Carolina Cristina
- Laboratorio de Fisiopatología de la Hipófisis; Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín B6000, Argentina; (L.R.); (C.C.)
| | - Marcela Fabiana Bolontrade
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB)-CONICET-Instituto Universitario del Hospital Italiano-Hospital Italiano Buenos Aires (HIBA), Buenos Aires C1199ACL, Argentina;
| | - Laura Alaniz
- Laboratorio de Microambiente Tumoral; Centro de Investigaciones Básicas y Aplicadas (CIBA), CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As. Consejo Nacional de Investigaciones Científicas y Técnicas (UNNOBA-CONICET), Junín B6000, Argentina; (I.S.); (F.M.S.); (D.L.V.); (A.I.)
| |
Collapse
|
15
|
When polymers meet carbon nanostructures: expanding horizons in cancer therapy. Future Med Chem 2020; 11:2205-2231. [PMID: 31538523 DOI: 10.4155/fmc-2018-0540] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The development of hybrid materials, which combine inorganic with organic materials, is receiving increasing attention by researchers. As a consequence of carbon nanostructures high chemical versatility, they exhibit enormous potential for new highly engineered multifunctional nanotherapeutic agents for cancer therapy. Whereas many groups are working on drug delivery systems for chemotherapy, the use of carbon nanohybrids for radiotherapy is rarely applied. Thus, nanotechnology offers a wide range of solutions to overcome the current obstacles of conventional chemo- and/or radiotherapies. Within this review, the structure and properties of carbon nanostructures (carbon nanotubes, nanographene oxide) functionalized preferentially with different types of polymers (synthetic, natural) are discussed. In short, synthesis approaches, toxicity investigations and anticancer efficacy of different carbon nanohybrids are described.
Collapse
|
16
|
Gadag S, Sinha S, Nayak Y, Garg S, Nayak UY. Combination Therapy and Nanoparticulate Systems: Smart Approaches for the Effective Treatment of Breast Cancer. Pharmaceutics 2020; 12:E524. [PMID: 32521684 PMCID: PMC7355786 DOI: 10.3390/pharmaceutics12060524] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer has become one of the biggest concerns for oncologists in the past few decades because of its unpredictable etiopathology and nonavailability of personalized translational medicine. The number of women getting affected by breast cancer has increased dramatically, owing to lifestyle and environmental changes. Besides, the development of multidrug resistance has become a challenge in the therapeutic management of breast cancer. Studies reveal that the use of monotherapy is not effective in the management of breast cancer due to high toxicity and the development of resistance. Combination therapies, such as radiation therapy with adjuvant therapy, endocrine therapy with chemotherapy, and targeted therapy with immunotherapy, are found to be effective. Thus, multimodal and combination treatments, along with nanomedicine, have emerged as a promising strategy with minimum side effects and drug resistance. In this review, we emphasize the multimodal approaches and recent advancements in breast cancer treatment modalities, giving importance to the current data on clinical trials. The novel treatment approach by targeted therapy, according to type, such as luminal, HER2 positive, and triple-negative breast cancer, are discussed. Further, passive and active targeting technologies, including nanoparticles, bioconjugate systems, stimuli-responsive, and nucleic acid delivery systems, including siRNA and aptamer, are explained. The recent research exploring the role of nanomedicine in combination therapy and the possible use of artificial intelligence in breast cancer therapy is also discussed herein. The complexity and dynamism of disease changes require the constant upgrading of knowledge, and innovation is essential for future drug development for treating breast cancer.
Collapse
Affiliation(s)
- Shivaprasad Gadag
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| | - Shristi Sinha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Sanjay Garg
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Usha Y. Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India; (S.G.); (S.S.)
| |
Collapse
|
17
|
Gao Y, Tang M, Leung E, Svirskis D, Shelling A, Wu Z. Dual or multiple drug loaded nanoparticles to target breast cancer stem cells. RSC Adv 2020; 10:19089-19105. [PMID: 35518295 PMCID: PMC9054075 DOI: 10.1039/d0ra02801k] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/13/2020] [Indexed: 12/18/2022] Open
Abstract
Breast cancer stem(-like) cells (BCSCs) have been found to be responsible for therapeutic resistance and disease relapse. BCSCs are difficult to eradicate due to their high resistance to conventional treatments and high plasticity. Functionalised nanoparticles have been investigated as smart vehicles to transport across various barriers and increase the interaction of therapeutic agents with cancer cells, as well as BCSCs. In this review, we discuss the different characteristics of BCSCs, and challenges to tackle BCSCs at cellular and molecular levels. The mechanisms of action and physicochemical properties of the current BCSC targeting agents are also covered. We will focus on the rational design and recent advances of "Nano + Nano" or single tumour targeting nanoparticle systems loaded with dual or multiple agents to kill all cancer cells including BCSCs. These cocktail therapies include the combination of a chemotherapy agent with a BCSC-specific inhibitor, a phytochemical agent or RNA based therapy. Given the heterogeneity of breast tumour tissue, targeting both BCSCs and bulk breast cancer cells simultaneously with multiple agents holds great promise in eliminating breast cancer. The future research needs to focus on overcoming various barriers in the 'clinical translation' of BCSC-targeting nanomedicines to cure breast cancer, which requires a significant multidisciplinary effort.
Collapse
Affiliation(s)
- Yu Gao
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland Auckland 1142 New Zealand +64-9-9231709
| | - Mingtan Tang
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland Auckland 1142 New Zealand +64-9-9231709
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland Auckland 1023 New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland Auckland 1142 New Zealand +64-9-9231709
| | - Andrew Shelling
- School of Medicine, Faculty of Medical and Health Sciences, The University of Auckland Auckland 1142 New Zealand
| | - Zimei Wu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland Auckland 1142 New Zealand +64-9-9231709
| |
Collapse
|
18
|
Wang S, Zhu X, Han M, Hao F, Lu W, Zhou T. Mechanistic Pharmacokinetic/Pharmacodynamic Model of Sunitinib and Dopamine in MCF-7/Adr Xenografts: Linking Cellular Heterogeneity to Tumour Burden. AAPS JOURNAL 2020; 22:45. [PMID: 32043246 DOI: 10.1208/s12248-020-0428-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/26/2020] [Indexed: 01/08/2023]
Abstract
The self-renewal and differentiation of cancer stem-like cells (CSCs) leads to cellular heterogeneity, causing one of the greatest challenges in cancer therapy. Growing evidence suggests that CSC-targeting therapy enhances the effect of concomitant antitumour therapy. To gain an in-depth understanding of this enhanced effect, the kinetic profile of estimated CSC frequency (the fraction of CSCs in tumour) was evaluated for in vivo characterization of cellular heterogeneity using sunitinib and dopamine as a paradigm combination therapy. Female MCF-7/Adr xenografted Balb/c nude mice were treated with sunitinib (p.o., 20 mg/kg) and dopamine (i.p., 50 mg/kg), alone or in combination. Estimated CSC frequency and tumour size were measured over time. Mechanistic PK/PD modelling was performed to quantitatively describe the relationship between drug concentration, estimated CSC frequency and tumour size. Sunitinib reduced tumour size by inducing apoptosis of differentiated tumour cells (DTCs) and enriched CSCs by stimulating its proliferation. Dopamine exhibited anti-CSC effects by suppressing the capacity of CSCs and inducing its differentiation. Simulation and animal studies indicated that concurrent administration was superior to sequential administration under current experimental conditions. Alongside tumour size, the current study provides mechanistic insights into the estimation of CSC frequency as an indicator for cellular heterogeneity. This forms the conceptual basis for in vivo characterization of other combination therapies in preclinical cancer studies.
Collapse
Affiliation(s)
- Siyuan Wang
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,Center for Precision Medicine Multi-Omics Research, Peking University Health Science Center, Beijing, 100191, China
| | - Xiao Zhu
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,Otago Pharmacometrics Group, School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Mengyi Han
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China
| | - Fangran Hao
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China
| | - Wei Lu
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Beijing, 100191, China
| | - Tianyan Zhou
- Department of Pharmaceutics, School of Pharmaceutical sciences, Peking University, Beijing, 100191, China.,State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Beijing, 100191, China
| |
Collapse
|
19
|
Chen C, Tian R, Zeng Y, Chu C, Liu G. Activatable Fluorescence Probes for “Turn-On” and Ratiometric Biosensing and Bioimaging: From NIR-I to NIR-II. Bioconjug Chem 2020; 31:276-292. [DOI: 10.1021/acs.bioconjchem.9b00734] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Chuan Chen
- Department of Pharmacy, Xiamen Medical College, Xiamen, Fujian 361023, China
| | - Rui Tian
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Yun Zeng
- Department of Pharmacy, Xiamen Medical College, Xiamen, Fujian 361023, China
| | - Chengchao Chu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine School of Public Health, Xiamen University, Xiamen, Fujian 361102, 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, Fujian 361102, China
| |
Collapse
|
20
|
|
21
|
Aminated Graphene Oxide as a Potential New Therapy for Colorectal Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3738980. [PMID: 31015889 PMCID: PMC6446092 DOI: 10.1155/2019/3738980] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/17/2018] [Accepted: 02/03/2019] [Indexed: 01/05/2023]
Abstract
Nanotechnology-based drug delivery systems for cancer therapy are the topic of interest for many researchers and scientists. Graphene oxide (GO) and its derivates are among the most extensively studied delivery systems of this type. The increased surface area, elevated loading capacity, and aptitude for surface functionalization together with the ability to induce reactive oxygen species make GO a promising tool for the development of novel anticancer therapies. Moreover, GO nanoparticles not only function as effective drug carriers but also have the potential to exert their own inhibitory effects on tumour cells. Recent results show that the functionalization of GO with different functional groups, namely, with amine groups, leads to increased reactivity of the nanoparticles. The last steers different hypotheses for the mechanisms through which this functionalization of GO could potentially lead to improved anticancer capacity. In this research, we have evaluated the potential of amine-functionalized graphene oxide nanoparticles (GO-NH2) as new molecules for colorectal cancer therapy. For the purpose, we have assessed the impact of aminated graphene oxide (GO) sheets on the viability of colon cancer cells, their potential to generate ROS, and their potential to influence cellular proliferation and survival. In order to elucidate their mechanism of action on the cellular systems, we have probed their genotoxic and cytostatic properties and compared them to pristine GO. Our results revealed that both GO samples (pristine and aminated) were composed of few-layer sheets with different particle sizes, zeta potential, and surface characteristics. Furthermore, we have detected increased cyto- and genotoxicity of the aminated GO nanoparticles following 24-hour exposure on Colon 26 cells. The last leads us to conclude that exposure of cancer cells to GO, namely, aminated GO, can significantly contribute to cancer cell killing by enhancing the cytotoxicity effect exerted through the induction of ROS, subsequent DNA damage, and apoptosis.
Collapse
|
22
|
Turdo A, Veschi V, Gaggianesi M, Chinnici A, Bianca P, Todaro M, Stassi G. Meeting the Challenge of Targeting Cancer Stem Cells. Front Cell Dev Biol 2019; 7:16. [PMID: 30834247 PMCID: PMC6387961 DOI: 10.3389/fcell.2019.00016] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022] Open
Abstract
Notwithstanding cancer patients benefit from a plethora of therapeutic alternatives, drug resistance remains a critical hurdle. Indeed, the high mortality rate is associated with metastatic disease, which is mostly incurable due to the refractoriness of metastatic cells to current treatments. Increasing data demonstrate that tumors contain a small subpopulation of cancer stem cells (CSCs) able to establish primary tumor and metastasis. CSCs are endowed with multiple treatment resistance capabilities comprising a highly efficient DNA damage repair machinery, the activation of survival pathways, enhanced cellular plasticity, immune evasion and the adaptation to a hostile microenvironment. Due to the presence of distinct cell populations within a tumor, cancer research has to face the major challenge of targeting the intra-tumoral as well as inter-tumoral heterogeneity. Thus, targeting molecular drivers operating in CSCs, in combination with standard treatments, may improve cancer patients’ outcomes, yielding long-lasting responses. Here, we report a comprehensive overview on the most significant therapeutic advances that have changed the known paradigms of cancer treatment with a particular emphasis on newly developed compounds that selectively affect the CSC population. Specifically, we are focusing on innovative therapeutic approaches including differentiation therapy, anti-angiogenic compounds, immunotherapy and inhibition of epigenetic enzymes and microenvironmental cues.
Collapse
Affiliation(s)
- Alice Turdo
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Aurora Chinnici
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Paola Bianca
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of PROMISE, University of Palermo, Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| |
Collapse
|
23
|
Shaik AS, Shaik AP, Bammidi VK, Al Faraj A. Effect of polyethylene glycol surface charge functionalization of SWCNT on the in vitro and in vivo nanotoxicity and biodistribution monitored noninvasively using MRI. Toxicol Mech Methods 2019; 29:233-243. [PMID: 30480460 DOI: 10.1080/15376516.2018.1540674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The current study evaluated in vitro and in vivo toxicity of carboxyl or amine polyethylene glycol (PEG) surface functionalization of single-walled carbon nanotubes (SWCNTs). Assessments of cytotoxicity, genotoxicity, immunotoxicity, and oxidative stress were performed in vitro and in vivo (in a 1-month follow-up study). The SWCNT biodistribution was investigated using noninvasive magnetic resonance imaging (MRI). Results confirmed the enhanced biocompatibility of PEG-functionalized SWCNTs compared to non-functionalized materials with significant decreases (p < 0.05) in the percentage of cell viability and increases in ROS generation, mitochondrial membrane potential, cell apoptosis, oxidative stress generation, and oxidative DNA damage in vitro. PEG-functionalized SWCNTs with amine terminals were found to induce prominent increases in ROS generation, mitochondrial membrane potential, and oxidative stress compared to carboxy functionalization. No significant difference in the biodistribution of either functionalized SWCNTs was observed in MRI. In vivo assessments revealed a statistically significant increase (p < 0.05) in oxidative stress as early as 24 h in serum and liver; however, all values normalized at 2 weeks' investigation time point. DNA damage was minimal with either PEG-COOH or PEG-NH2 functionalized SWCNTs after 2 weeks' exposure. The negatively charged SWCNTs caused lesser DNA damage compared to positively charged samples. Carboxy-functionalized SWCNTs did not cause substantial changes in inflammatory mediators and were found to be significantly safer than non-functionalized SWCNTs and may pave the way for novel biomedical applications in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Asma Sultana Shaik
- a Prince Naif Health Research Center , King Saud University , Riyadh , Saudi Arabia
| | - Abjal Pasha Shaik
- b Department of Clinical Lab Sciences , College of Applied Medical Sciences, King Saud University , Riyadh , Saudi Arabia
| | - Vamsee K Bammidi
- c The Unicare Group, Burton-on-Trent , Staffordshire , United Kingdom
| | - Achraf Al Faraj
- d Department of Radiologic Sciences, Faculty of Health Science , American University of Science and Technology , Beirut , Lebanon
| |
Collapse
|
24
|
Sun Y, Kim HS, Kang S, Piao YJ, Jon S, Moon WK. Magnetic Resonance Imaging-Guided Drug Delivery to Breast Cancer Stem-Like Cells. Adv Healthc Mater 2018; 7:e1800266. [PMID: 30146770 DOI: 10.1002/adhm.201800266] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/14/2018] [Indexed: 01/16/2023]
Abstract
The feasibility of detecting breast cancer stem-like cells (BCSCs) with magnetic resonance imaging using extradomain-B of fibronectin (EDB-FN)-specific peptide (APTEDB )-conjugated thermally cross-linked superparamagnetic iron oxide nanoparticles (APTEDB -TCL-SPIONs) is previously demonstrated. Here, doxorubicin (Dox)-loaded APTEDB -TCL-SPIONs (Dox@APTEDB -TCL-SPIONs) are generated and their theranostic ability in a BCSC xenograft mouse model is assessed. The Dox@APTEDB -TCL-SPIONs enable more efficient delivery of Dox to tumors than nontargeted Dox@TCL-SPIONs. Much greater inhibition of BCSC tumor growth is observed after treatment with the Dox@APTEDB -TCL-SPIONs than with either Dox@TCL-SPIONs or free Dox. Hypointense signals are observed in the majority of the mice in postcontrast but not precontrast T2*-weighted MR images of tumors 7 days after treatment with Dox@APTEDB -TCL-SPIONs. An inverse correlation is observed between signal intensity and both EDB-FN expression and response to chemotherapy. The data indicate Dox@APTEDB -TCL-SPIONs can detect BCSCs within tumors by targeting EDB-FN-expressing cells. These nanoparticles thus have theranostic potential in breast cancer.
Collapse
Affiliation(s)
- Yujin Sun
- Department of Radiology; Seoul National University Hospital; 101 Daehak-ro Jongno-gu Seoul 03080 Republic of Korea
- Department of Radiology; Yanbian University Hospital; 1327 JuZi Street Yanji City Jilin Province 133000 China
| | - Hoe Suk Kim
- Department of Radiology; Seoul National University Hospital; 101 Daehak-ro Jongno-gu Seoul 03080 Republic of Korea
| | - Sukmo Kang
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); 291 Daehak-ro Yuseong-gu Daejeon 34141 Republic of Korea
| | - Yin Ji Piao
- Department of Biomedical Science and Radiology; Seoul National University College of Medicine; 103 Daehak-ro Jongno-gu Seoul 03080 Republic of Korea
| | - Sangyong Jon
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); 291 Daehak-ro Yuseong-gu Daejeon 34141 Republic of Korea
| | - Woo Kyung Moon
- Department of Radiology; Seoul National University Hospital; 101 Daehak-ro Jongno-gu Seoul 03080 Republic of Korea
- Department of Biomedical Science and Radiology; Seoul National University College of Medicine; 103 Daehak-ro Jongno-gu Seoul 03080 Republic of Korea
| |
Collapse
|
25
|
Mocan T, Matea CT, Pop T, Mosteanu O, Buzoianu AD, Suciu S, Puia C, Zdrehus C, Iancu C, Mocan L. Carbon nanotubes as anti-bacterial agents. Cell Mol Life Sci 2017; 74:3467-3479. [PMID: 28536787 PMCID: PMC11107489 DOI: 10.1007/s00018-017-2532-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/19/2017] [Accepted: 04/27/2017] [Indexed: 01/01/2023]
Abstract
Multidrug-resistant bacterial infections that have evolved via natural selection have increased alarmingly at a global level. Thus, there is a strong need for the development of novel antibiotics for the treatment of these infections. Functionalized carbon nanotubes through their unique properties hold great promise in the fight against multidrug-resistant bacterial infections. This new family of nanovectors for therapeutic delivery proved to be innovative and efficient for the transport and cellular translocation of therapeutic molecules. The current review examines the latest progress in the antibacterial activity of carbon nanotubes and their composites.
Collapse
Affiliation(s)
- Teodora Mocan
- Department of Nanomedicine, "Octavian Fodor" Gastroenterology Institute, 19-21 Croitorilor Street, Cluj-Napoca, Romania
- Department of Physiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 3-5 Clinicilor Street, Cluj-Napoca, Romania
| | - Cristian T Matea
- Department of Nanomedicine, "Octavian Fodor" Gastroenterology Institute, 19-21 Croitorilor Street, Cluj-Napoca, Romania
| | - Teodora Pop
- 3rd Gastroenterology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, 19-21 Croitorilor Street, Cluj-Napoca, Romania
| | - Ofelia Mosteanu
- 3rd Gastroenterology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy, 19-21 Croitorilor Street, Cluj-Napoca, Romania
| | - Anca Dana Buzoianu
- Department of Clinical Pharmacology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 3-5 Clinicilor Street, Cluj-Napoca, Romania
| | - Soimita Suciu
- Department of Physiology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 3-5 Clinicilor Street, Cluj-Napoca, Romania
| | - Cosmin Puia
- Department of Nanomedicine, "Octavian Fodor" Gastroenterology Institute, 19-21 Croitorilor Street, Cluj-Napoca, Romania
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, 19-21 Croitorilor Street, Cluj-Napoca, Romania
| | - Claudiu Zdrehus
- Department of Nanomedicine, "Octavian Fodor" Gastroenterology Institute, 19-21 Croitorilor Street, Cluj-Napoca, Romania
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, 19-21 Croitorilor Street, Cluj-Napoca, Romania
| | - Cornel Iancu
- Department of Nanomedicine, "Octavian Fodor" Gastroenterology Institute, 19-21 Croitorilor Street, Cluj-Napoca, Romania.
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, 19-21 Croitorilor Street, Cluj-Napoca, Romania.
| | - Lucian Mocan
- Department of Nanomedicine, "Octavian Fodor" Gastroenterology Institute, 19-21 Croitorilor Street, Cluj-Napoca, Romania.
- 3rd Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy, 19-21 Croitorilor Street, Cluj-Napoca, Romania.
| |
Collapse
|
26
|
Sheikhpour M, Golbabaie A, Kasaeian A. Carbon nanotubes: A review of novel strategies for cancer diagnosis and treatment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:1289-1304. [DOI: 10.1016/j.msec.2017.02.132] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 12/18/2016] [Accepted: 02/24/2017] [Indexed: 12/25/2022]
|
27
|
Sanginario A, Miccoli B, Demarchi D. Carbon Nanotubes as an Effective Opportunity for Cancer Diagnosis and Treatment. BIOSENSORS 2017; 7:E9. [PMID: 28212271 PMCID: PMC5371782 DOI: 10.3390/bios7010009] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/21/2022]
Abstract
Despite the current progresses of modern medicine, the resistance of malignant tumors to present medical treatments points to the necessity of developing new therapeutic approaches. In recent years, numerous studies have focused their attention on the promising use of nanomaterials, like iron oxide nanowires, zinc oxide or mesoporous silica nanoparticles, for cancer and metastasis treatment with the advantage of operating directly at the bio-molecular scale. Among them, carbon nanotubes emerged as valid candidates not only for drug delivery, but also as a valuable tool in cancer imaging and physical ablation. Nevertheless, deep investigations about carbon nanotubes' potential bio-compatibility and cytotoxicity limits should be also critically addressed. In the present review, after introducing carbon nanotubes and their promising advantages and drawbacks for fighting cancer, we want to focus on the numerous and different ways in which they can assist to reach this goal. Specifically, we report on how they can be used not only for drug delivery purposes, but also as a powerful ally to develop effective contrast agents for tumors' medical or photodynamic imaging, to perform direct physical ablation of metastasis, as well as gene therapy.
Collapse
Affiliation(s)
- Alessandro Sanginario
- Electronics Design Laboratory (EDL), Istituto Italiano di Tecnologia, Via Melen 83b, 16152 Genova (GE), Italy.
| | - Beatrice Miccoli
- Department of Electronics and Telecommunications, Politecnico di Torino, C.so Duca degli Abruzzi 24, 10129 Torino, Italy.
| | - Danilo Demarchi
- Department of Electronics and Telecommunications, Politecnico di Torino, C.so Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
28
|
Qin W, Huang G, Chen Z, Zhang Y. Nanomaterials in Targeting Cancer Stem Cells for Cancer Therapy. Front Pharmacol 2017; 8:1. [PMID: 28149278 PMCID: PMC5241315 DOI: 10.3389/fphar.2017.00001] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/03/2017] [Indexed: 12/25/2022] Open
Abstract
Cancer stem cells (CSCs) have been identified in almost all cancers and give rise to metastases and can also act as a reservoir of cancer cells that may cause a relapse after surgery, radiation, or chemotherapy. Thus they are obvious targets in therapeutic approaches and also a great challenge in cancer treatment. The threat presented by CSCs lies in their unlimited proliferative ability and multidrug resistance. These findings have necessitated an effective novel strategy to target CSCs for cancer treatment. Nanomaterials are on the route to providing novel methods in cancer therapies. Although, there have been a large number of excellent work in the field of targeted cancer therapy, it remains an open question how nanomaterials can meet future demands for targeting and eradicating of CSCs. In this review, we summarized recent and highlighted future prospects for targeting CSCs for cancer therapies by using a variety of nanomaterials.
Collapse
Affiliation(s)
- Weiwei Qin
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| | - Guan Huang
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| | - Zuanguang Chen
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| | - Yuanqing Zhang
- Institute of Medical Instrument and Application, School of Pharmaceutical Sciences, Sun Yat-Sen University Guangzhou, China
| |
Collapse
|
29
|
Augustine S, Singh J, Srivastava M, Sharma M, Das A, Malhotra BD. Recent advances in carbon based nanosystems for cancer theranostics. Biomater Sci 2017; 5:901-952. [DOI: 10.1039/c7bm00008a] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review deals with four different types of carbon allotrope based nanosystems and summarizes the results of recent studies that are likely to have applications in cancer theranostics. We discuss the applications of these nanosystems for cancer imaging, drug delivery, hyperthermia, and PDT/TA/PA.
Collapse
Affiliation(s)
- Shine Augustine
- NanoBioelectronics Laboratory
- Department of Biotechnology
- Delhi Technological University
- Delhi 110042
- India
| | - Jay Singh
- Department of Applied Chemistry & Polymer Technology
- Delhi Technological University
- Delhi 110042
- India
| | - Manish Srivastava
- Department of Physics & Astrophysics
- University of Delhi
- Delhi 110007
- India
| | - Monica Sharma
- NanoBioelectronics Laboratory
- Department of Biotechnology
- Delhi Technological University
- Delhi 110042
- India
| | - Asmita Das
- NanoBioelectronics Laboratory
- Department of Biotechnology
- Delhi Technological University
- Delhi 110042
- India
| | - Bansi D. Malhotra
- NanoBioelectronics Laboratory
- Department of Biotechnology
- Delhi Technological University
- Delhi 110042
- India
| |
Collapse
|
30
|
Pan J, Li F, Choi JH. Single-walled carbon nanotubes as optical probes for bio-sensing and imaging. J Mater Chem B 2017; 5:6511-6522. [PMID: 32264414 DOI: 10.1039/c7tb00748e] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A review on the applications of single-walled carbon nanotube photoluminescence in biomolecular sensing and biomedical imaging.
Collapse
Affiliation(s)
- Jing Pan
- School of Mechanical Engineering
- Purdue University
- West Lafayette
- USA
| | - Feiran Li
- School of Mechanical Engineering
- Purdue University
- West Lafayette
- USA
| | - Jong Hyun Choi
- School of Mechanical Engineering
- Purdue University
- West Lafayette
- USA
| |
Collapse
|
31
|
Halwani R, Sultana Shaik A, Ratemi E, Afzal S, Kenana R, Al-Muhsen S, Al Faraj A. A novel anti-IL4Rα nanoparticle efficiently controls lung inflammation during asthma. Exp Mol Med 2016; 48:e262. [PMID: 27713399 PMCID: PMC5099422 DOI: 10.1038/emm.2016.89] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/19/2016] [Accepted: 04/24/2016] [Indexed: 01/08/2023] Open
Abstract
Drug resistance and the harmful side effects accompanying the prolonged corticosteroid treatment of chronic pulmonary diseases prompted the development of more specific anti-inflammatory approaches. Several strategies aiming to block IL4Rα, the receptor for a key pro-inflammatory pathway, were investigated. However, their efficiency was limited, mostly due to the systemic or subcutaneous route of administrations. In this paper, we examined the ability of an intranasal treatment with biocompatible nanoparticles targeting IL4Rα to control lung inflammation in ovalbumin (OVA)-sensitized mice. OVA-sensitized mice were treated with anti-IL4Rα-conjugated nanoparticles. The levels of pro-inflammatory cytokines in the lungs and broncho-alveolar lavage fluid (BALF) were determined using a cytokine array assay. The effects of nanoparticle treatment on the activation of lung inflammatory cells and their ability to proliferate and produce cytokines were determined using fluorescence-activated cell sorting (FACS) analysis. Lung inflammation was also monitored using immunohistochemical staining. Treatment with the anti-IL4Rα nanoparticles significantly decreased pro-inflammatory cytokine expression and release in BALF and airway lung tissue in mice. The numbers of lung tissue lymphocytes, neutrophils and eosinophils were also decreased. Interestingly, anti-IL4Rα nanoparticles deactivated CD4 and CD8 T cells in lung tissue and inhibited their ability to produce pro-inflammatory cytokines to a significantly lower level than the treatment with free anti-IL4Rα. Moreover, they induced a sustained low level of lung inflammation for 1 week following the last instillation compared with the treatment with free anti-IL4Rα antibodies. Together, this data suggested that the enhanced tissue penetrability and sustainability of these nanoparticles improved the strength and durability of the immunosuppressive effects of anti-IL4Rα.
Collapse
Affiliation(s)
- Rabih Halwani
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Asma Sultana Shaik
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Prince Naif Health Research Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Elaref Ratemi
- Jubail Industrial College, Department of Chemical and Process Engineering Technology, Jubail Industrial City, Saudi Arabia
| | - Sibtain Afzal
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Rosan Kenana
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Prince Naif Health Research Center, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Saleh Al-Muhsen
- Prince Naif Center for Immunology Research and Asthma Research Chair, Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Achraf Al Faraj
- Department of Radiological Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
32
|
Hernández-Rivera M, Zaibaq NG, Wilson LJ. Toward carbon nanotube-based imaging agents for the clinic. Biomaterials 2016; 101:229-40. [DOI: 10.1016/j.biomaterials.2016.05.045] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/12/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022]
|
33
|
Shaik AP, Shaik AS, Majwal AA, Faraj AA. Blocking Interleukin-4 Receptor α Using Polyethylene Glycol Functionalized Superparamagnetic Iron Oxide Nanocarriers to Inhibit Breast Cancer Cell Proliferation. Cancer Res Treat 2016; 49:322-329. [PMID: 27456946 PMCID: PMC5398403 DOI: 10.4143/crt.2016.091] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/16/2016] [Indexed: 12/25/2022] Open
Abstract
Purpose The specific targeting of interleukin-4 receptor α (IL4Rα) receptor offers a promising therapeutic approach for inhibition of tumor cell progression in breast cancer patients. In the current study, the in vitro efficacy of superparamagnetic iron oxide nanoparticles conjugated with anti-IL4Rα blocking antibodies (SPION-IL4Rα) via polyethylene glycol polymers was evaluated in 4T1 breast cancer cells. Materials and Methods Cell viability, reactive oxygen species generation, and apoptosis frequency were assessed in vitro in 4T1 cancer cell lines following exposure to SPION-IL4Rα alone or combined with doxorubicin. In addition, immunofluorescence assessments and fluorimetrywere performed to confirm the specific targeting and interaction of the developed nanocarriers with IL4Rα receptors in breast cancer cells. Results Blocking of IL4Rα receptors caused a significant decrease in cell viability and induced apoptosis in 4T1 cells. In addition, combined treatment with SPION-IL4Rα+doxorubicin caused significant increases in cell death, apoptosis, and oxidative stress compared to either SPION-IL4Rα or doxorubicin alone, indicating the enhanced therapeutic efficacy of this combination. The decrease in fluorescence intensity upon immunofluorescence and fluorimetry assays combined with increased viability and decreased apoptosis following the blocking of IL4Rα receptors confirmed the successful binding of the synthesized nanocarriers to the target sites on murine 4T1 breast cancerous cells. Conclusion These results suggest that SPION-IL4Rα nanocarriers might be used for successfulreduction of tumor growth and inhibition of progression of metastasis in vivo.
Collapse
Affiliation(s)
- Abjal Pasha Shaik
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Asma Sultana Shaik
- Prince Naif Health Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ali Al Majwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Achraf Al Faraj
- Department of Radiological Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Al Faraj A. SWCNTs as novel theranostic nanocarriers for cancer diagnosis and therapy: towards safe translation to the clinics. Nanomedicine (Lond) 2016; 11:1431-45. [DOI: 10.2217/nnm-2016-0065] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
With their unique physicochemical properties, single walled carbon nanotubes (SWCNTs) hold great promise for applications as drug delivery systems (DDS) for early and better diagnosis and therapy of cancer. While several in vitro and in vivo studies have validated their potential benefit, no SWCNT-based formulation has yet reached clinical trials. Towards prospective safe clinical applications, the main properties that were adopted to enhance the biocompatibility of SWCNTs were highlighted. Then, the recent progresses in the in vivo applications of SWCNTs as diagnostic nanoprobes using multimodality imaging techniques and as therapeutic nanocarriers delivering wide range of anticancer efficient drugs to tumors were reviewed. Finally, the efforts required for safe clinical applications of SWCNTs as DDS for cancer diagnosis and therapy were discussed.
Collapse
Affiliation(s)
- Achraf Al Faraj
- Department of Radiological Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
35
|
Wang C, Li Y, Chen B, Zou M. In vivo pharmacokinetics, biodistribution and the anti-tumor effect of cyclic RGD-modified doxorubicin-loaded polymers in tumor-bearing mice. Colloids Surf B Biointerfaces 2016; 146:31-8. [PMID: 27244048 DOI: 10.1016/j.colsurfb.2016.05.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/14/2016] [Accepted: 05/17/2016] [Indexed: 11/17/2022]
Abstract
In our previous study, we successfully produced and characterized a multifunctional drug delivery system with doxorubicin (RC/GO/DOX), which was based on graphene oxide (GO) and cyclic RGD-modified chitosan (RC). Its characteristics include: pH-responsiveness, active targeting of hepatocarcinoma cells, and efficient loading with controlled drug release. Here, we report the pharmacokinetics, biodistribution, and anti-tumor efficacy of RC/GO/DOX polymers in tumor-bearing nude mice. The objective of this study is to assess its targeting potential for tumors. Pharmacokinetic and biodistribution profiles demonstrated that tumor accumulation of RC/GO/DOX polymers was almost three times higher than the others, highlighting the efficacy of the active targeting strategy. Furthermore, the tumor inhibition rate of RC/GO/DOX polymers was 56.64%, 2.09 and 2.93 times higher than that of CS/GO/DOX polymers (without modification) and the DOX solution, respectively. Anti-tumor efficacy results indicated that the tumor growth was better controlled by RC/GO/DOX polymers than the others. Hematoxylin and eosin (H&E) staining showed remarkable changes in tumor histology. Compared with the saline group, the tumor section from the RC/GO/DOX group revealed a marked increase in the quantity of apoptotic and necrotic cells, and a reduction in the quantity of the blood vessels. Together, these studies show that this new system could be regarded as a suitable form of DOX-based treatment of the hepatocellular carcinoma.
Collapse
Affiliation(s)
- Chen Wang
- School of Pharmacy, Xiamen Medical College, Xiamen 361008, PR China.
| | - Yuan Li
- School of Pharmacy, Xiamen Medical College, Xiamen 361008, PR China
| | - Binbin Chen
- Department of Pharmacy, Xiamen Xianyue Hospital, 361012, PR China
| | - Meijuan Zou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| |
Collapse
|
36
|
Al Faraj A, Shaik AS, Ratemi E, Halwani R. Combination of drug-conjugated SWCNT nanocarriers for efficient therapy of cancer stem cells in a breast cancer animal model. J Control Release 2016; 225:240-51. [PMID: 26827662 DOI: 10.1016/j.jconrel.2016.01.053] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/24/2016] [Accepted: 01/27/2016] [Indexed: 01/06/2023]
Abstract
Targeting breast cancer and more specifically cancer stem cell (CSC) subpopulation, responsible for tumor growth, resistance and self-renewal, using combination of therapeutic drugs selectively delivered via biocompatible nanocarriers, provides a novel approach for effective therapy. Here, we propose to evaluate the potential therapeutic efficacy of combining Paclitaxel and Salinomycin drugs actively targeted to both breast cancer and CSCs in xenograft murine model after conjugation with biocompatible CD44 antibody conjugated SWCNTs via hydrazone linker allowing pH-responsive release mechanism near the acidic tumor microenvironment. Both in vitro investigations on MDA-MB-231, sorted CSC negative or CSC positive fractions and in vivo evaluations on tumor-bearing mice using noninvasive bioluminescence and magnetic resonance imaging confirmed the enhanced therapeutic effect of the combined therapy compared to treatment with individual drug-conjugated nanocarriers or free drug suspensions. Thus, confirmed the great promise of the developed SWCNTs drug delivery system for effective breast cancer treatment by targeting and eradicating both whole tumor cells and CSCs populations.
Collapse
Affiliation(s)
- Achraf Al Faraj
- King Saud University, College of Applied Medical Sciences, Department of Radiological Sciences, Riyadh, Saudi Arabia.
| | - Asma Sultana Shaik
- King Saud University, Prince Naif Health Research Center, Riyadh, Saudi Arabia
| | - Elaref Ratemi
- Jubail Industrial College, Department of Chemical and Process Engineering Technology, Jubail Industrial City, Saudi Arabia
| | - Rabih Halwani
- King Saud University, College of Medicine, Prince Naif Center for Immunology Research, Department of Pediatrics, Riyadh, Saudi Arabia
| |
Collapse
|