1
|
Sueyoshi S, Vitor Silva J, Guizze F, Giarolla J. Dendrimers as drug delivery systems for oncotherapy: Current status of promising applications. Int J Pharm 2024; 663:124573. [PMID: 39134292 DOI: 10.1016/j.ijpharm.2024.124573] [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: 02/19/2024] [Revised: 08/08/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Cancer affects millions of people worldwide, causing death and serious health problems. Despite significant investment in the development of new anticancer compounds, there are still several limitations that can still be found. Many compounds exhibit high levels of toxicity and low bioavailability. Therefore, it is urgent to design safer, more effective, and particularly more selective compounds for oncological treatment. Dendrimers are polymeric structures that have been shown to be potential drug nanocarriers to overcome physicochemical, pharmacokinetic, and indirect pharmacodynamic issues. Due to their versatility, they can be used in the design of nanovaccines, lipophilic complexes, amphiphilic complexes, smart nanocomplexes, and others. This work targets the use of dendrimers in oncological treatment and their importance and effectiveness as drug delivery systems for the development of new therapies. For this review, only publications from the last two years are considered in this review.
Collapse
Affiliation(s)
- Sophia Sueyoshi
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - João Vitor Silva
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - Felipe Guizze
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil
| | - Jeanine Giarolla
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, Av Prof Lineu Prestes, 580, Bl. 13, CEP 05508-900 São Paulo, SP, Brazil.
| |
Collapse
|
2
|
Rodríguez-Candela Mateos M, Carpintero-Fernández P, Freijanes PS, Mosquera J, Nebril BA, Mayán MD. Insights into the role of connexins and specialized intercellular communication pathways in breast cancer: Mechanisms and applications. Biochim Biophys Acta Rev Cancer 2024; 1879:189173. [PMID: 39154967 DOI: 10.1016/j.bbcan.2024.189173] [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: 05/27/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Gap junctions, membrane-based channels comprised of connexin proteins (Cxs), facilitate direct communication among neighbouring cells and between cells and the extracellular space through their hemichannels. The normal human breast expresses various Cxs family proteins, such as Cx43, Cx30, Cx32, Cx46, and Cx26, crucial for proper tissue development and function. These proteins play a significant role in breast cancer development, progression, and therapy response. In primary tumours, there is often a reduction and cytoplasmic mislocalization of Cx43 and Cx26, while metastatic lesions show an upregulation of these and other Cxs. Although existing research predominantly supports the tumour-suppressing role of Cxs in primary carcinomas through channel-dependent and independent functions, controversies persist regarding their involvement in the metastatic process. This review aims to provide an updated perspective on Cxs in human breast cancer, with a specific focus on intrinsic subtypes due to the heterogeneous nature of this disease. Additionally, the manuscript will explore the role of Cxs in immune interactions and novel forms of intercellular communication, such as tunneling nanotubes and extracellular vesicles, within the breast tumour context and tumour microenvironment. Recent findings suggest that Cxs hold potential as therapeutic targets for mitigating metastasis and drug resistance. Furthermore, they may serve as novel biomarkers for cancer prognosis, offering promising avenues for future research and clinical applications.
Collapse
Affiliation(s)
- Marina Rodríguez-Candela Mateos
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain
| | - Paula Carpintero-Fernández
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; CellCOM Research Group, Center for Research in Nanomaterials and Biomedicine (CINBIO), Universidade de Vigo, Edificio Olimpia Valencia, Campus Universitario Lagoas Marcosende, 36310 Vigo, Spain; Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS, Spain
| | - Paz Santiago Freijanes
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Anatomic Pathology Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - Joaquin Mosquera
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Surgery Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - Benigno Acea Nebril
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Surgery Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - María D Mayán
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; CellCOM Research Group, Center for Research in Nanomaterials and Biomedicine (CINBIO), Universidade de Vigo, Edificio Olimpia Valencia, Campus Universitario Lagoas Marcosende, 36310 Vigo, Spain; Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS, Spain.
| |
Collapse
|
3
|
Batool Z, Kamal MA, Shen B. Advancements in triple-negative breast cancer sub-typing, diagnosis and treatment with assistance of artificial intelligence : a focused review. J Cancer Res Clin Oncol 2024; 150:383. [PMID: 39103624 PMCID: PMC11300496 DOI: 10.1007/s00432-024-05903-2] [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: 05/07/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Triple negative breast cancer (TNBC) is most aggressive type of breast cancer with multiple invasive sub-types and leading cause of women's death worldwide. Lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2) causes it to spread rapidly making its treatment challenging due to unresponsiveness towards anti-HER and endocrine therapy. Hence, needing advanced therapeutic treatments and strategies in order to get better recovery from TNBC. Artificial intelligence (AI) has been emerged by giving its high inputs in the automated diagnosis as well as treatment of several diseases, particularly TNBC. AI based TNBC molecular sub-typing, diagnosis as well as therapeutic treatment has become successful now days. Therefore, present review has reviewed recent advancements in the role and assistance of AI particularly focusing on molecular sub-typing, diagnosis as well as treatment of TNBC. Meanwhile, advantages, certain limitations and future implications of AI assistance in the TNBC diagnosis and treatment are also discussed in order to fully understand readers regarding this issue.
Collapse
Affiliation(s)
- Zahra Batool
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610218, China
| | - Mohammad Amjad Kamal
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610218, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Bairong Shen
- Center for High Altitude Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610218, China.
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, A-10, No.17, Tianfu Avenue, Shangliu Distinct, Chengdu, 610002, China.
| |
Collapse
|
4
|
Saharkhiz S, Nasri N, Naderi N, Dini G, Ghalehshahi SS, Firoozbakht F. Evaluating a targeted Palbociclib-Trastuzumab loaded smart niosome platform for treating HER2 positive breast cancer cells. Int J Pharm X 2024; 7:100237. [PMID: 38516198 PMCID: PMC10955288 DOI: 10.1016/j.ijpx.2024.100237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
In this study, we present a targeted and pH-sensitive niosomal (pHSN) formulation, incorporating quantum dot (QD)-labeled Trastuzumab (Trz) molecules for the specific delivery of Palbociclib (Pal) to cells overexpressing human epidermal growth factor receptor 2 (HER2). FTIR analyses confirmed the successful preparation of the pHSNs and their bioconjugation. The labeled Trz-conjugated Pal-pHSNs (Trz-Pal-pHSNs) exhibited a size of approximately 170 nm, displaying a spherical shape with a neutral surface charge of -1.2 mV. Pal encapsulation reached ∼86%, and the release pattern followed a two-phase pH-dependent mechanism. MTT assessments demonstrated enhanced apoptosis induction, particularly in HER2-positive cells, by Trz-Pal-pHSNs. Fluorescence imaging further validated the internalization of particles into cells. In conclusion, Trz-Pal-pHSNs emerge as a promising platform for personalized medicine in the treatment of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Shaghayegh Saharkhiz
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Negar Nasri
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Nazanin Naderi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran 19839-69411, Iran
| | - Ghasem Dini
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| | - Saeid Shirzadi Ghalehshahi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 81746-73441, Iran
| | - Fateme Firoozbakht
- Department of Nanotechnology, Faculty of Chemistry, University of Isfahan, Isfahan 81746-73441, Iran
| |
Collapse
|
5
|
Binmujlli MA. Radiological and Molecular Analysis of Radioiodinated Anastrozole and Epirubicin as Innovative Radiopharmaceuticals Targeting Methylenetetrahydrofolate Dehydrogenase 2 in Solid Tumors. Pharmaceutics 2024; 16:616. [PMID: 38794278 PMCID: PMC11126143 DOI: 10.3390/pharmaceutics16050616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic field of radiopharmaceuticals, innovating targeted agents for cancer diagnosis and therapy is crucial. Our study enriches this evolving landscape by evaluating the potential of radioiodinated anastrozole ([125I]anastrozole) and radioiodinated epirubicin ([125I]epirubicin) as targeting agents against MTHFD2-driven tumors. MTHFD2, which is pivotal in one-carbon metabolism, is notably upregulated in various cancers, presenting a novel target for radiopharmaceutical application. Through molecular docking and 200 ns molecular dynamics (MD) simulations, we assess the binding efficiency and stability of [125I]anastrozole and [125I]epirubicin with MTHFD2. Molecular docking illustrates that [125I]epirubicin has a superior binding free energy (∆Gbind) of -41.25 kJ/mol compared to -39.07 kJ/mol for [125I]anastrozole and -38.53 kJ/mol for the control ligand, suggesting that it has a higher affinity for MTHFD2. MD simulations reinforce this, showing stable binding, as evidenced by root mean square deviation (RMSD) values within a narrow range, underscoring the structural integrity of the enzyme-ligand complexes. The root mean square fluctuation (RMSF) analysis indicates consistent dynamic behavior of the MTHFD2 complex upon binding with [125I]anastrozole and [125I]epirubicin akin to the control. The radius of gyration (RG) measurements of 16.90 Å for MTHFD2-[125I]anastrozole and 16.84 Å for MTHFD2-[125I]epirubicin confirm minimal structural disruption upon binding. The hydrogen bond analysis reveals averages of two and three stable hydrogen bonds for [125I]anastrozole and [125I]epirubicin complexes, respectively, highlighting crucial stabilizing interactions. The MM-PBSA calculations further endorse the thermodynamic favorability of these interactions, with binding free energies of -48.49 ± 0.11 kJ/mol for [125I]anastrozole and -43.8 kJ/mol for MTHFD2-. The significant contribution of Van der Waals and electrostatic interactions to the binding affinities of [125I]anastrozole and [125I]epirubicin, respectively, underscores their potential efficacy for targeted tumor imaging and therapy. These computational findings lay the groundwork for the future experimental validation of [125I]anastrozole and [125I]epirubicin as MTHFD2 inhibitors, heralding a notable advancement in precision oncology tools. The data necessitate subsequent in vitro and in vivo assays to corroborate these results.
Collapse
Affiliation(s)
- Mazen Abdulrahman Binmujlli
- Department of Internal Medicine, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh 11623, Saudi Arabia
| |
Collapse
|
6
|
Fernandes NB, Velagacherla V, Spandana KJ, N B, Mehta CH, Gadag S, Sabhahit JN, Nayak UY. Co-delivery of lapatinib and 5-fluorouracil transfersomes using transpapillary iontophoresis for breast cancer therapy. Int J Pharm 2024; 650:123686. [PMID: 38070658 DOI: 10.1016/j.ijpharm.2023.123686] [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: 07/05/2023] [Revised: 11/18/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023]
Abstract
Combination chemotherapy, involving the intervention of two or more anti-neoplastic agents has been the cornerstone in breast cancer treatment, owing to the applications it holds in contrast to the mono-therapy approach. This research predominantly focussed on proving the synergy between Lapatinib (LPT) and 5-Fluorouracil (5-FU) and further enhancing its localized permeation via transfersome-loaded delivery and iontophoresis to treat breast tumors. The IC50 values for LPT and 5-FU were found to be 19.38 µg/ml and 5.7 µg/ml respectively and their synergistic effect was proven by the Chou-Talalay assay using CompuSyn software. Furthermore, LPT and 5-FU were encapsulated within transfersomes and administered via the transpapillary route. The drug-loaded carriers were characterized for their particle size, polydispersity index, zeta potential, and entrapment efficiency. The ex vivo rat skin permeation studies indicated that when compared to LPT dispersion and 5-FU solution, drug-loaded transfersomes exhibited better permeability and their transpapillary permeation was enhanced on using iontophoresis. Moreover, both LPT and 5-FU transfersomes were found to be stable for 3 months when stored at a temperature of 5 ± 3 °C. The results indicated that this treatment strategy could be an effective approach in contrast to some of the conventional treatments employed to date.
Collapse
Affiliation(s)
- Neha B Fernandes
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Varalakshmi Velagacherla
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - K J Spandana
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru 575018, Karnataka, India
| | - Bhagya N
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangaluru 575018, Karnataka, India
| | - Chetan H Mehta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shivaprasad Gadag
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Jayalakshmi N Sabhahit
- Department of Electrical and Electronics Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
7
|
Guo J, Hu J, Zheng Y, Zhao S, Ma J. Artificial intelligence: opportunities and challenges in the clinical applications of triple-negative breast cancer. Br J Cancer 2023; 128:2141-2149. [PMID: 36871044 PMCID: PMC10241896 DOI: 10.1038/s41416-023-02215-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for 15-20% of all invasive breast cancer subtypes. Owing to its clinical characteristics, such as the lack of effective therapeutic targets, high invasiveness, and high recurrence rate, TNBC is difficult to treat and has a poor prognosis. Currently, with the accumulation of large amounts of medical data and the development of computing technology, artificial intelligence (AI), particularly machine learning, has been applied to various aspects of TNBC research, including early screening, diagnosis, identification of molecular subtypes, personalised treatment, and prediction of prognosis and treatment response. In this review, we discussed the general principles of artificial intelligence, summarised its main applications in the diagnosis and treatment of TNBC, and provided new ideas and theoretical basis for the clinical diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Jiamin Guo
- Department of Medical Oncology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan Province, P. R. China
| | - Junjie Hu
- Machine Intelligence Laboratory, College of Computer Science, Sichuan University, 610065, Chengdu, Sichuan Province, P. R. China
| | - Yichen Zheng
- Department of Medical Oncology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan Province, P. R. China
| | - Shuang Zhao
- Department of Radiology, West China Hospital of Sichuan University, 610041, Chengdu, Sichuan Province, P. R. China.
| | - Ji Ma
- Department of Medical Oncology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan Province, P. R. China.
| |
Collapse
|
8
|
Mehta A. Tracking the Development of Cancer Care After 75 Years of Independence: India's Fight Against Cancer Since 1947. Indian J Surg Oncol 2022; 13:12-26. [PMID: 36691502 PMCID: PMC9859970 DOI: 10.1007/s13193-022-01689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
India is one of the fastest developing countries with tremendous growth in industrialization and healthcare facilities. Research and development in the field of healthcare improved the quality of life and well-being of our population. Despite the availability of healthcare facilities and infrastructure, we are still facing considerable challenges in the prevention, diagnosis, and treatment of cancer. The present review focuses on the history and development of cancer care facilities since independence. The advances in cancer diagnostics for early detection of cancer and developments in the field of conventional surgery, including laparoscopic and robotic surgeries, chemotherapy, and radiation therapy, are reviewed. Immunotherapy, vaccines, and selective targeting of tumor cells using nanotechnology are emerging areas in the field of cancer research.
Collapse
Affiliation(s)
- Ashok Mehta
- Nanavati Max Super Speciality Hospital, Mumbai, India
- L S Raheja Hospital, Mumbai, India
- HCG Cancer Centre Colaba, Mumbai, India
| |
Collapse
|
9
|
Gadag S, Narayan R, Sabhahit JN, Hari G, Nayak Y, Pai KSR, Garg S, Nayak UY. Transpapillary iontophoretic delivery of resveratrol loaded transfersomes for localized delivery to breast cancer. BIOMATERIALS ADVANCES 2022; 140:213085. [PMID: 36037762 DOI: 10.1016/j.bioadv.2022.213085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/29/2022] [Accepted: 08/14/2022] [Indexed: 06/15/2023]
Abstract
Localized drug delivery to the breast tissues is an area of interest as a potential route to ensure site-specific drug delivery. Transpapillary delivery via the mammary papilla has advantages as most breast tumors arise from the milk ducts. The present study explored the plausibility of transpapillary delivery of a phytochemical, resveratrol (RVT), for breast cancer treatment. RVT was encapsulated within the transfersomes (RVT-TRF) to enable a sustained release of the drug using the biomaterial soya phosphatidylcholine (SPC). Iontophoresis was applied to further accelerate the penetration of the RVT-TRF across the mammary papilla to the breast tissue. The RVT-TRF development was optimized by the Design of Experiments (DoE) approach. The in vitro transpapillary iontophoresis study on porcine mammary papilla showed an enhanced penetration of RVT-TRF when compared to passive diffusion. The transpapillary delivery was further confirmed from the in vitro fluorescent microscopy study using FITC conjugated RVT-TRF. The optimized RVT-TRF delivered via transpapillary route showed a higher Cmax and AUC when compared to pure RVT given orally. A significant reduction in the tumor volume and the serum biomarker CA 15-3, when evaluated in a chemically induced breast cancer rat model, provided evidence of the effectiveness of the developed formulation when delivered locally via transpapillary route compared to the oral route. Thus the developed RVT-TRF administered via transpapillary iontophoresis technique is a promising strategy enabling a localized delivery for effective breast cancer therapy.
Collapse
Affiliation(s)
- Shivaprasad Gadag
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Reema Narayan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Jayalakshmi N Sabhahit
- Department of Electrical and Electronics Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Gangadhar Hari
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Karkala Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, 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, Karnataka, India.
| |
Collapse
|
10
|
Zhou L, Zou M, Xu Y, Lin P, Lei C, Xia X. Nano Drug Delivery System for Tumor Immunotherapy: Next-Generation Therapeutics. Front Oncol 2022; 12:864301. [PMID: 35664731 PMCID: PMC9160744 DOI: 10.3389/fonc.2022.864301] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Abstract
Tumor immunotherapy is an artificial stimulation of the immune system to enhance anti-cancer response. It has become a powerful clinical strategy for treating cancer. The number of immunotherapy drug approvals has been increasing in recent years, and many treatments are in clinical and preclinical stages. Despite this progress, the special tumor heterogeneity and immunosuppressive microenvironment of solid tumors made immunotherapy in the majority of cancer cases difficult. Therefore, understanding how to improve the intratumoral enrichment degree and the response rate of various immunotherapy drugs is key to improve efficacy and control adverse reactions. With the development of materials science and nanotechnology, advanced biomaterials such as nanoparticle and drug delivery systems like T-cell delivery therapy can improve effectiveness of immunotherapy while reducing the toxic side effects on non-target cells, which offers innovative ideas for improving immunity therapeutic effectiveness. In this review, we discuss the mechanism of tumor cell immune escape and focus on current immunotherapy (such as cytokine immunotherapy, therapeutic monoclonal antibody immunotherapy, PD-1/PD-L1 therapy, CAR-T therapy, tumor vaccine, oncolytic virus, and other new types of immunity) and its challenges as well as the latest nanotechnology (such as bionic nanoparticles, self-assembled nanoparticles, deformable nanoparticles, photothermal effect nanoparticles, stimuli-responsive nanoparticles, and other types) applications in cancer immunotherapy.
Collapse
Affiliation(s)
- Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Manshu Zou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yilin Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Peng Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Chang Lei
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
11
|
Khakinahad Y, Sohrabi S, Razi S, Narmani A, Khaleghi S, Asadiyun M, Jafari H, Mohammadnejad J. Margetuximab conjugated-PEG-PAMAM G4 nano-complex: a smart nano-device for suppression of breast cancer. Biomed Eng Lett 2022; 12:317-329. [PMID: 35892030 PMCID: PMC9308845 DOI: 10.1007/s13534-022-00225-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 01/06/2022] [Accepted: 03/06/2022] [Indexed: 12/18/2022] Open
Abstract
Abstract Breast cancer due to its high incidence and mortality is the second leading cause of death among females. On the other hand, nanoparticle-based drug delivery is one of the most promising approaches in cancer therapy, nowadays. Hence, margetuximab- and polyethylene glycol-conjugated PAMAM G4 dendrimers were efficiently synthesized for targeted delivery of quercetin (therapeutic agent) to MDA-MB-231 breast cancer cells. Synthesized nano-complexes were characterized using analytical devices such as FT-IR, TGA, DLS, Zeta potential analyzer, and TEM. The size less than 40 nm, - 18.8 mV surface charge, efficient drug loading capacity (21.48%), and controlled drug release (about 45% of drug release normal pH after 8 h) were determined for the nano-complex. In the biomedical test, the cell viability was obtained 14.67% at 24 h of post-treatment for 800 nM concentration, and IC50 was ascertained at 100 nM for the nano-complex. The expression of apoptotic Bax and Caspase9 genes was increased by more than eightfolds and more than fivefolds after treatment with an optimal concentration of nanocarrier. Also, more than threefolds of cell cycle arrest was observed at the optimal concentration synthetics, and 27.5% breast cancer cell apoptosis was detected after treatment with 100 nM nano-complex. These outputs have been indicating the potential capacity of synthesized nano-complex in inhibiting the growth of breast cancer cells. Graphic abstract
Collapse
Affiliation(s)
- Yasaman Khakinahad
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
| | - Saeedeh Sohrabi
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
- Department of Biology, Faculty of Advanced Sciences and Technology, Payam Noor University, Tehran, Iran
| | - Shokufeh Razi
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Sepideh Khaleghi
- Department of Medical Biotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahboubeh Asadiyun
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Department of Biological and Biomedical Sciences, Cancer Biomedical Center, Tehran, Iran
| | - Hanieh Jafari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Javad Mohammadnejad
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
12
|
Ribociclib-Loaded Ethylcellulose-Based Nanosponges: Formulation, Physicochemical Characterization, and Cytotoxic Potential against Breast Cancer. ADSORPT SCI TECHNOL 2022. [DOI: 10.1155/2022/1922263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In the present study, ribociclib-loaded nanosponges (RCNs) composed of ethylcellulose and polyvinyl alcohol were developed using an emulsion-solvent evaporation method. Preliminary evaluations of the developed RCNs (RCN1 to RCN7) were performed in terms of size, polydispersity index (PDI), zeta potential (ZP), entrapment efficiency (EE), and drug loading (DL), which allowed us to select the optimized formulation. RCN3 was selected as the optimized carrier system with particle size (
), PDI (
), zeta potential (
), EE (
), and DL (
). Further, the optimized nanosponges (RCN3) were subjected to FTIR, XRD, DSC, and SEM studies, and results confirmed the proper encapsulation of the drug within the porous polymeric matrix. In vitro drug release studies showed that the drug release was significantly enhanced with a maximum drug release through RCN3 formulation (
) and followed the Higuchi model. Moreover, the RCN3 system showed greater cytotoxicity than free ribociclib (RC) against MDA-MB-231 and MCF-7 breast cancer cell lines. The percentage of apoptosis induced by RCN3 was found significantly higher than that of free RC (
). Overall, ribociclib-loaded ethylcellulose nanosponges could be a potential nanocarrier to enhance the effectiveness of ribociclib in breast cancer treatment.
Collapse
|
13
|
Noda N, Ozawa T. Castanospermine suppresses CD44 ectodomain cleavage as revealed by transmembrane bioluminescent sensors. J Cell Sci 2022; 135:274740. [PMID: 35194645 DOI: 10.1242/jcs.259314] [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: 08/24/2021] [Accepted: 02/12/2022] [Indexed: 11/20/2022] Open
Abstract
Cluster of differentiation 44 (CD44) is a single-pass transmembrane glycoprotein that is a widely distributed cell-surface adhesion molecule. CD44 undergoes ectodomain cleavage by membrane-associated metalloproteinases in breast cancer cells. Cleavage plays a critical role in cancer cell migration by mediating the interaction between CD44 and the extracellular matrix. To explore inhibitors of CD44 ectodomain cleavage, we developed two bioluminescent sensors for the detection of CD44 ectodomain cleavage. The sensors were designed as two-transmembrane proteins with split-luciferase fragments, one of which was cyclized by protein trans-splicing of a DnaE intein. These two sensors emit light by the cyclization or the spontaneous complementation of the luciferase fragments. The luminescence intensities decreased by cleavage of the ectodomain in breast cancer cells. The sensors revealed that castanospermine, an α-glucosidase inhibitor, suppressed the ectodomain cleavage of endogenous CD44 in breast cancer cells. Castanospermine also inhibited breast cancer cell invasion. Thus, the sensors are beneficial tools for evaluating the effects of different inhibitors.
Collapse
Affiliation(s)
- Natsumi Noda
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeaki Ozawa
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
14
|
St-Denis-Bissonnette F, Khoury R, Mediratta K, El-Sahli S, Wang L, Lavoie JR. Applications of Extracellular Vesicles in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:451. [PMID: 35053616 PMCID: PMC8773485 DOI: 10.3390/cancers14020451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and refractory subtype of breast cancer, often occurring in younger patients with poor clinical prognosis. Given the current lack of specific targets for effective intervention, the development of better treatment strategies remains an unmet medical need. Over the last decade, the field of extracellular vesicles (EVs) has grown tremendously, offering immense potential for clinical diagnosis/prognosis and therapeutic applications. While TNBC-EVs have been shown to play an important role in tumorigenesis, chemoresistance and metastasis, they could be repurposed as potential biomarkers for TNBC diagnosis and prognosis. Furthermore, EVs from various cell types can be utilized as nanoscale drug delivery systems (NDDS) for TNBC treatment. Remarkably, EVs generated from specific immune cell subsets have been shown to delay solid tumour growth and reduce tumour burden, suggesting a new immunotherapy approach for TNBC. Intrinsically, EVs can cross the blood-brain barrier (BBB), which holds great potential to treat the brain metastases diagnosed in one third of TNBC patients that remains a substantial clinical challenge. In this review, we present the most recent applications of EVs in TNBC as diagnostic/prognostic biomarkers, nanoscale drug delivery systems and immunotherapeutic agents, as well as discuss the associated challenges and future directions of EVs in cancer immunotherapy.
Collapse
Affiliation(s)
- Frederic St-Denis-Bissonnette
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Rachil Khoury
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Karan Mediratta
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Jessie R. Lavoie
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada; (F.S.-D.-B.); (R.K.); (K.M.); (S.E.-S.)
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| |
Collapse
|
15
|
Rizwanullah M, Ahmad MZ, Ghoneim MM, Alshehri S, Imam SS, Md S, Alhakamy NA, Jain K, Ahmad J. Receptor-Mediated Targeted Delivery of Surface-ModifiedNanomedicine in Breast Cancer: Recent Update and Challenges. Pharmaceutics 2021; 13:2039. [PMID: 34959321 PMCID: PMC8708551 DOI: 10.3390/pharmaceutics13122039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer therapeutic intervention continues to be ambiguous owing to the lack of strategies for targeted transport and receptor-mediated uptake of drugs by cancer cells. In addition to this, sporadic tumor microenvironment, prominent restrictions with conventional chemotherapy, and multidrug-resistant mechanisms of breast cancer cells possess a big challenge to even otherwise optimal and efficacious breast cancer treatment strategies. Surface-modified nanomedicines can expedite the cellular uptake and delivery of drug-loaded nanoparticulate constructs through binding with specific receptors overexpressed aberrantly on the tumor cell. The present review elucidates the interesting yet challenging concept of targeted delivery approaches by exploiting different types of nanoparticulate systems with multiple targeting ligands to target overexpressed receptors of breast cancer cells. The therapeutic efficacy of these novel approaches in preclinical models is also comprehensively discussed in this review. It is concluded from critical analysis of related literature that insight into the translational gap between laboratories and clinical settings would provide the possible future directions to plug the loopholes in the process of development of these receptor-targeted nanomedicines for the treatment of breast cancer.
Collapse
Affiliation(s)
- Md. Rizwanullah
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia;
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.)
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.)
| | - Keerti Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)—Raebareli, Lucknow 226002, India;
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia;
| |
Collapse
|
16
|
Gadag S, Narayan R, Nayak AS, Catalina Ardila D, Sant S, Nayak Y, Garg S, Nayak UY. Development and preclinical evaluation of microneedle-assisted resveratrol loaded nanostructured lipid carriers for localized delivery to breast cancer therapy. Int J Pharm 2021; 606:120877. [PMID: 34252522 PMCID: PMC8429179 DOI: 10.1016/j.ijpharm.2021.120877] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/08/2021] [Accepted: 07/07/2021] [Indexed: 01/08/2023]
Abstract
Resveratrol (RVT) is one of the potent anticancer phytochemicals which has shown promising potential for breast cancer therapy. However, its short half-life and low bioavailability is a major hurdle in its effective use. In this study, we have developed nanostructured lipid carriers (NLCs) of RVT to enable localized delivery of the drug to the breast tissues using microneedle arrays to improve effectiveness. The NLCs were optimized using the Design of Experiments approach and characterized for their particle size, polydispersity index, zeta potential and entrapment efficiency. The RVT-NLCs delivered using microneedle array 1200 showed a higher permeation of RVT across the skin with lower skin retention compared to pure RVT. Further, RVT-NLCs showed higher anticancer activity on MDA-MB-231 breast cancer cell lines and enhanced internalization compared to pure RVT. Moreover, the RVT-NLCs were found to inhibit the migration of MDA-MB-231 breast cancer cell lines. Preclinical studies in rats showed that RVT-NLCs delivered via microneedles demonstrated a remarkable increase in the Cmax, Tmax and AUC0-inf, and a higher localization in breast tissue compared to pure RVT administered orally. These results suggests that the RVT-NLCs administered by microneedle array system is an effective strategy for the local delivery of RVT for breast cancer therapy.
Collapse
Affiliation(s)
- Shivaprasad Gadag
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Reema Narayan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Archana S Nayak
- Department of Chemical Engineering, National Institute of Technology Karnataka, Surathkal, Mangalore, Karnataka, 575025, India
| | - Diana Catalina Ardila
- Department of Pharmaceutical Sciences, School of Pharmacy, Pittsburgh, PA 15261, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, Pittsburgh, PA 15261, USA; Department of Bioengineering, Swanson School of Engineering, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15261, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15261, USA
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 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, Karnataka 576104, India.
| |
Collapse
|
17
|
Bădilă AE, Rădulescu DM, Niculescu AG, Grumezescu AM, Rădulescu M, Rădulescu AR. Recent Advances in the Treatment of Bone Metastases and Primary Bone Tumors: An Up-to-Date Review. Cancers (Basel) 2021; 13:4229. [PMID: 34439383 PMCID: PMC8392383 DOI: 10.3390/cancers13164229] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/14/2021] [Accepted: 08/20/2021] [Indexed: 12/13/2022] Open
Abstract
In the last decades, the treatment of primary and secondary bone tumors has faced a slow-down in its development, being mainly based on chemotherapy, radiotherapy, and surgical interventions. However, these conventional therapeutic strategies present a series of disadvantages (e.g., multidrug resistance, tumor recurrence, severe side effects, formation of large bone defects), which limit their application and efficacy. In recent years, these procedures were combined with several adjuvant therapies, with different degrees of success. To overcome the drawbacks of current therapies and improve treatment outcomes, other strategies started being investigated, like carrier-mediated drug delivery, bone substitutes for repairing bone defects, and multifunctional scaffolds with bone tissue regeneration and antitumor properties. Thus, this paper aims to present the types of bone tumors and their current treatment approaches, further focusing on the recent advances in new therapeutic alternatives.
Collapse
Affiliation(s)
- Adrian Emilian Bădilă
- “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.E.B.); (D.M.R.); (A.R.R.)
- Department of Orthopedics and Traumatology, Bucharest University Hospital, 050098 Bucharest, Romania
| | - Dragoș Mihai Rădulescu
- “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.E.B.); (D.M.R.); (A.R.R.)
- Department of Orthopedics and Traumatology, Bucharest University Hospital, 050098 Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania; (A.-G.N.); (A.M.G.)
| | - Alexandru Mihai Grumezescu
- Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania; (A.-G.N.); (A.M.G.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 50044 Bucharest, Romania
| | - Marius Rădulescu
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, University Politehnica of Bucharest, 1-7 Polizu St., 011061 Bucharest, Romania
| | - Adrian Radu Rădulescu
- “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.E.B.); (D.M.R.); (A.R.R.)
- Department of Orthopedics and Traumatology, Bucharest University Hospital, 050098 Bucharest, Romania
| |
Collapse
|
18
|
Exploring the therapeutic potential of nanostructured lipid carrier approaches to tackling the inherent lacuna of chemotherapeutics and herbal drugs against breast cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
19
|
Iron Oxide-Based Magneto-Optical Nanocomposites for In Vivo Biomedical Applications. Biomedicines 2021; 9:biomedicines9030288. [PMID: 34156393 PMCID: PMC8000024 DOI: 10.3390/biomedicines9030288] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 01/07/2023] Open
Abstract
Iron oxide nanoparticles (IONPs) have played a pivotal role in the development of nanomedicine owing to their versatile functions at the nanoscale, which facilitates targeted delivery, high contrast imaging, and on-demand therapy. Some biomedical inadequacies of IONPs on their own, such as the poor resolution of IONP-based Magnetic Resonance Imaging (MRI), can be overcome by co-incorporating optical probes onto them, which can be either molecule- or nanoparticulate-based. Optical probe incorporated IONPs, together with two prominent non-ionizing radiation sources (i.e., magnetic field and light), enable a myriad of biomedical applications from early detection to targeted treatment of various diseases. In this context, many research articles are in the public domain on magneto-optical nanoparticles; discussed in detail are fabrication strategies for their application in the biomedical field; however, lacking is a comprehensive review on real-life applications in vivo, their toxicity, and the prospect of bench-to-bedside clinical studies. Therefore, in this review, we focused on selecting such important nanocomposites where IONPs become the magnetic component, conjugated with various types of optical probes; we clearly classified them into class 1 to class 6 categories and present only in vivo studies. In addition, we briefly discuss the potential toxicity of such nanocomposites and their respective challenges for clinical translations.
Collapse
|
20
|
Wu Y, Li M, Lin J, Hu C. Hippo/TEAD4 signaling pathway as a potential target for the treatment of breast cancer. Oncol Lett 2021; 21:313. [PMID: 33692845 PMCID: PMC7933775 DOI: 10.3892/ol.2021.12574] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/03/2021] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most common type of cancer among women worldwide. The Hippo signaling pathway is strongly associated with cell proliferation, migration, invasion, metastasis and resistance to breast cancer treatment. The upstream factors involved in the Hippo signaling pathway, including mammalian Ste20 kinases 1/2, large tumor suppressor kinases 1/2 and transcription coactivator Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ), have been extensively studied as they are considered therapeutic targets for breast cancer. Recently, it has been suggested that the transcriptional enhancer factor domain (TEAD) family of transcription factors, particularly TEAD4, plays an important role in breast cancer. TEADs interact with YAP/TAZ to act as transcription factors. Notably, recent studies have demonstrated that TEAD4 may also function in a YAP/TAZ-independent manner and serve as a prognostic marker for breast cancer. The present review summarizes the current research on the effect of the aberrant activation of the Hippo signaling pathway on breast cancer progression. Furthermore, the latest advances on the role of the TEAD family in breast cancer are highlighted, and the role of TEAD4 as a potential target for therapeutic intervention in breast cancer is discussed.
Collapse
Affiliation(s)
- Yujian Wu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Mengjie Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Jiayi Lin
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Chenxia Hu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
21
|
Concept Design, Development and Preliminary Physical and Chemical Characterization of Tamoxifen-Guided-Mesoporous Silica Nanoparticles. Molecules 2021; 26:molecules26010219. [PMID: 33406699 PMCID: PMC7795496 DOI: 10.3390/molecules26010219] [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: 10/27/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Conventional chemotherapies used for breast cancer (BC) treatment are non-selective, attacking both healthy and cancerous cells. Therefore, new technologies that enhance drug efficacy and ameliorate the off-target toxic effects exhibited by currently used anticancer drugs are urgently needed. Here we report the design and synthesis of novel mesoporous silica nanoparticles (MSNs) equipped with the hormonal drug tamoxifen (TAM) to facilitate guidance towards estrogen receptors (ERs) which are upregulated in breast tumours. TAM is linked to the MSNs using a poly-ʟ-histidine (PLH) polymer as a pH-sensitive gatekeeper, to ensure efficient delivery of encapsulated materials within the pores. XRD, HR-TEM, DLS, SEM, FT-IR and BET techniques were used to confirm the successful fabrication of MSNs. The MSNs have a high surface area (>1000 m2/g); and a mean particle size of 150 nm, which is an appropriate size to allow the penetration of premature blood vessels surrounding breast tumours. Successful surface functionalization was supported by FT-IR, XPS and TGA techniques, with a grafting ratio of approximately 29%. The outcomes of this preliminary work could be used as practical building blocks towards future formulations.
Collapse
|