1
|
Fobian SF, Amin M, Sacchetti A, Oei AL, Ten Hagen TLM. Investigating the delivery of PD-L1-targeted immunoliposomes in a dynamic cervical cancer-on-a-chip model. J Control Release 2025; 379:236-250. [PMID: 39798703 DOI: 10.1016/j.jconrel.2025.01.014] [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: 06/30/2024] [Revised: 11/14/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The recent approval of pembrolizumab in recurrent or metastatic cervical cancer warrants further investigations into the usefulness of immunotherapies for more durable and less radical interventions. In this study, the targeting potential of anti-PD-L1-functionalized immunoliposomes was tested in a 3D in vitro cervical cancer-on-a-chip model. Immunolipsomes were synthesized and decorated externally with monovalent anti-PD-L1 Fab' fragments of commercially available atezolizumab. Cervical cancer cell lines with varying levels of PD-L1 expression were cultured as spheroids embedded in a collagen I matrix, and treated under flow of culture media. Flow cytometry and live-cell confocal imaging were used to measure the interactions and uptake of untargeted liposomes and immunoliposomes in this panel of cell lines. The immunoliposomes retained specific functionality regardless of protein corona formation in high serum environments. As such, spheroids expressing high levels of PD-L1 preferentially internalized immunoliposomes in a 3D environment with extracellular matrix present, while low PD-L1-expressing cell lines showed no preference for either formulation. Importantly, treatments performed in monolayer cultures (on plastic) showed no differences between immuno- and untargeted liposome uptake, including the way in which the endocytosed liposomes are trafficked subcellularly. This study demonstrates the importance of both active and passive accumulation strategies to achieve nanoparticle targeting. Immunoliposomes remain a promising platform for the development of targeted nanotherapies against cervical cancers. However, initial functional tests did not translate directly to biological performance and this should be kept in mind for future formulations. Furthermore, the in vitro model developed appeared useful for visualizing liposome uptake in a 3D, live tissue environment and represents a cost-effective and reproducible model for future studies.
Collapse
Affiliation(s)
- Seth-Frerich Fobian
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands; Department of Radiation Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Cancer Center Amsterdam (CCA), Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Mohamadreza Amin
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Andrea Sacchetti
- Department of Pathology, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Arlene L Oei
- Department of Radiation Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Cancer Center Amsterdam (CCA), Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands.
| |
Collapse
|
2
|
Gabr BS, Shalabi AR, Said MF, George RF. 3,5-Disubstituted pyrazoline as a promising core for anticancer agents: mechanisms of action and therapeutic potentials. Future Med Chem 2025; 17:725-745. [PMID: 40079157 PMCID: PMC11938987 DOI: 10.1080/17568919.2025.2476393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
The rapidly growing interest in the literature about the anticancer activity of 3,5-disubstituted pyrazolines and their promising therapeutic potentials/pharmacological properties, supported by the number of pyrazoline derivatives currently in clinical use or clinical trials, encouraged us to review the in vitro antiproliferative effects and biochemical investigations of probable mechanisms of action. Nevertheless, many reported pyrazoline-bearing compounds have anticancer activity without an explored mode of action, which opens new research avenues to examine their biochemical profiles further. Therefore, 3,5-disubstituted pyrazoline is a promising core that can be used to design new derivatives with anticancer activity based on the structure-activity relationship summarized in this review to obtain higher potency and selectivity.
Collapse
Affiliation(s)
- Basma S. Gabr
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sinai University, EL-Arish, Egypt
| | - Abdelrahman R. Shalabi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sinai University, EL-Arish, Egypt
| | - Mona F. Said
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Riham F. George
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
3
|
Radhakrishnan A, Shanmukhan NK, Samuel LC. Pharmacogenomics influence on MDR1-associated cancer resistance and innovative drug delivery approaches: advancing precision oncology. Med Oncol 2025; 42:67. [PMID: 39913003 DOI: 10.1007/s12032-025-02611-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025]
Abstract
Currently, there is a growing concern surrounding the treatment of cancer, a formidable disease. Pharmacogenomics and personalized medicine have emerged as significant areas of interest in cancer management. The efficacy of many cancer drugs is hindered by resistance mechanisms, particularly P-glycoprotein (P-gp) efflux, leading to reduced therapeutic outcomes. Efforts have intensified to inhibit P-gp efflux, thereby enhancing the effectiveness of resistant drugs. P-gp, a member of the ATP-binding cassette (ABC) superfamily, specifically the multidrug resistance (MDR)/transporter associated with antigen processing (TAP) sub-family B, member 1, utilizes energy derived from ATP hydrolysis to drive efflux. This review focuses on genetic polymorphisms associated with P-gp efflux and explores various novel pharmaceutical strategies to address this challenge. These strategies encompass SEDDS/SNEDDS, liposomes, immunoliposomes, solid lipid nanoparticles, lipid core nanocapsules, microemulsions, dendrimers, hydrogels, polymer-drug conjugates, and polymeric nanoparticles. The article aims to elucidate the interplay between pharmacogenomics, P-gp-mediated drug resistance in cancer, and formulation strategies to improve cancer therapy by tailoring formulations to genetically susceptible patients.
Collapse
Affiliation(s)
- Arun Radhakrishnan
- Department of Pharmaceutics, JKKN College of Pharmacy, Kumarapalayam, Tamil Nadu, 638183, India.
| | - Nikhitha K Shanmukhan
- Department of Pharmaceutics, JKKN College of Pharmacy, Kumarapalayam, Tamil Nadu, 638183, India
| | - Linda Christabel Samuel
- Department of Conservative Dentistry and Endodontics, JKKN Dental College and Hospitals, Kumarapalayam, 638183, India
| |
Collapse
|
4
|
Liao CC, Long Y, Tsai ML, Lin CY, Hsu KW, Lee CH. G-cleave LC3B biosensor: monitoring autophagy and assessing resveratrol's synergistic impact on doxorubicin-induced apoptosis in breast cancer cells. Breast Cancer Res 2024; 26:190. [PMID: 39736723 DOI: 10.1186/s13058-024-01951-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025] Open
Abstract
Autophagy, a crucial process in cancer, is closely intertwined with both tumor progression and drug resistance development. However, existing methods used to assess autophagy activity often pose invasiveness and time-related constraints, limiting their applicability in preclinical drug investigations. In this study, we developed a non-invasive autophagy detection system (NIADS-autophagy, also called G-cleave LC3B biosensor) by integrating a split-luciferase-based biosensor with an LC3B cleavage sequence, which swiftly identified classic autophagic triggers, such as Earle's Balanced Salt Solution and serum deprivation, through protease-mediated degradation pathways. The specificity of G-cleave LC3B biosensor was confirmed via CRISPR gene editing of pivotal autophagy regulator ATG4B, yielding diminished luciferase activity in MDA-MB-231 breast cancer cells. Notably, the G-cleave LC3B biosensor exhibited strong concordance with established autophagy metrics, encompassing LC3B lipidation, SQSTM1 degradation, and puncta accumulation analysis. To underscore the usage potential of the G-cleave LC3B biosensor, we discovered that resveratrol acts as a synergistic enhancer by significantly potentiating apoptosis in MDA-MB-231 cells when combined with doxorubicin treatment. Overall, the luminescence-based G-cleave LC3B biosensor presents a rapid and dependable avenue for determining autophagy activity, thereby facilitating high-throughput assessment of promising autophagy-associated anti-cancer therapies across diverse malignancies.
Collapse
Affiliation(s)
- Chiao-Chun Liao
- Department of Tropical Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City, Taiwan
| | - Yuqing Long
- Respiratory Medicine Unit and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Nuffield Department of Medicine Experimental Medicine, University of Oxford, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Ming-Lin Tsai
- Department of General Surgery, Cathay General Hospital, Taipei, Taiwan
| | - Chun-Yu Lin
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Kai-Wen Hsu
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung City, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung City, Taiwan
| | - Chia-Hwa Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City, Taiwan.
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
5
|
Mugundhan SL, Mohan M. Nanoscale strides: exploring innovative therapies for breast cancer treatment. RSC Adv 2024; 14:14017-14040. [PMID: 38686289 PMCID: PMC11056947 DOI: 10.1039/d4ra02639j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
Breast cancer (BC) is a predominant malignancy in women that constitutes approximately 30% of all cancer cases and has a mortality rate of 14% in recent years. The prevailing therapies include surgery, chemotherapy, and radiotherapy, each with its own limitations and challenges. Despite oral or intravenous administration, there are numerous barriers to accessing anti-BC agents before they reach the tumor site, including physical, physiological, and biophysical barriers. The complexity of BC pathogenesis, attributed to a combination of endogenous, chronic, intrinsic, extrinsic and genetic factors, further complicates its management. Due to the limitations of existing cancer treatment approaches, there is a need to explore novel, efficacious solutions. Nanodrug delivery has emerged as a promising avenue in cancer chemotherapy, aiming to enhance drug bioavailability while mitigating adverse effects. In contrast to conventional chemotherapy, cancer nanotechnology leverages improved permeability to achieve comprehensive disruption of cancer cells. This approach also presented superior pharmacokinetic profiles. The application of nanotechnology in cancer therapeutics includes nanotechnological tools, but a comprehensive review cannot cover all facets. Thus, this review concentrates specifically on BC treatment. The focus lies in the successful implementation of systematic nanotherapeutic strategies, demonstrating their superiority over conventional methods in delivering anti-BC agents. Nanotechnology-driven drug delivery holds immense potential in treating BC. By surmounting multiple barriers and capitalizing on improved permeability, nanodrug delivery has demonstrated enhanced efficacy and reduced adverse effects compared to conventional therapies. This review highlights the significance of systematic nanotherapy approaches, emphasizing the evolving landscape of BC management.
Collapse
Affiliation(s)
- Sruthi Laakshmi Mugundhan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology SRM Nagar Kattankulathur 603203 Tamil Nadu India
| | - Mothilal Mohan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology SRM Nagar Kattankulathur 603203 Tamil Nadu India
| |
Collapse
|
6
|
López-Estévez AM, Lapuhs P, Pineiro-Alonso L, Alonso MJ. Personalized Cancer Nanomedicine: Overcoming Biological Barriers for Intracellular Delivery of Biopharmaceuticals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309355. [PMID: 38104275 DOI: 10.1002/adma.202309355] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/09/2023] [Indexed: 12/19/2023]
Abstract
The success of personalized medicine in oncology relies on using highly effective and precise therapeutic modalities such as small interfering RNA (siRNA) and monoclonal antibodies (mAbs). Unfortunately, the clinical exploitation of these biological drugs has encountered obstacles in overcoming intricate biological barriers. Drug delivery technologies represent a plausible strategy to overcome such barriers, ultimately facilitating the access to intracellular domains. Here, an overview of the current landscape on how nanotechnology has dealt with protein corona phenomena as a first and determinant biological barrier is presented. This continues with the analysis of strategies facilitating access to the tumor, along with conceivable methods for enhanced tumor penetration. As a final step, the cellular barriers that nanocarriers must confront in order for their biological cargo to reach their target are deeply analyzed. This review concludes with a critical analysis and future perspectives of the translational advances in personalized oncological nanomedicine.
Collapse
Affiliation(s)
- Ana María López-Estévez
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Philipp Lapuhs
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Laura Pineiro-Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| |
Collapse
|
7
|
Li Y, Dong D, Qu Y, Li J, Chen S, Zhao H, Zhang Q, Jiao Y, Fan L, Sun D. A Multidrug Delivery Microrobot for the Synergistic Treatment of Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301889. [PMID: 37423966 DOI: 10.1002/smll.202301889] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/21/2023] [Indexed: 07/11/2023]
Abstract
Multidrug combination therapy provides an effective strategy for malignant tumor treatment. This paper presents the development of a biodegradable microrobot for on-demand multidrug delivery. By combining magnetic targeting transportation with tumor therapy, it is hypothesized that loading multiple drugs on different regions of a single magnetic microrobot can enhance a synergistic effect for cancer treatment. The synergistic effect of using two drugs together is greater than that of using each drug separately. Here, a 3D-printed microrobot inspired by the fish structure with three hydrogel components: skeleton, head, and body structures is demonstrated. Made of iron oxide (Fe3 O4 ) nanoparticles embedded in poly(ethylene glycol) diacrylate (PEGDA), the skeleton can respond to magnetic fields for microrobot actuation and drug-targeted delivery. The drug storage structures, head, and body, made by biodegradable gelatin methacryloyl (GelMA) exhibit enzyme-responsive cargo release. The multidrug delivery microrobots carrying acetylsalicylic acid (ASA) and doxorubicin (DOX) in drug storage structures, respectively, exhibit the excellent synergistic effects of ASA and DOX by accelerating HeLa cell apoptosis and inhibiting HeLa cell metastasis. In vivo studies indicate that the microrobots improve the efficiency of tumor inhibition and induce a response to anti-angiogenesis. The versatile multidrug delivery microrobot conceptualized here provides a way for developing effective combination therapy for cancer.
Collapse
Affiliation(s)
- Yanfang Li
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Dingran Dong
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Yun Qu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Junyang Li
- Center for Robotics and Automation, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518000, China
- Department of Electronic Engineering, Ocean University of China, Qingdao, 266000, China
| | - Shuxun Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Han Zhao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Qi Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Yang Jiao
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Lei Fan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
- Center for Robotics and Automation, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518000, China
| | - Dong Sun
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
- Center for Robotics and Automation, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518000, China
| |
Collapse
|
8
|
Lei Z, Tian Q, Teng Q, Wurpel JND, Zeng L, Pan Y, Chen Z. Understanding and targeting resistance mechanisms in cancer. MedComm (Beijing) 2023; 4:e265. [PMID: 37229486 PMCID: PMC10203373 DOI: 10.1002/mco2.265] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/05/2023] [Accepted: 03/23/2023] [Indexed: 05/27/2023] Open
Abstract
Resistance to cancer therapies has been a commonly observed phenomenon in clinical practice, which is one of the major causes of treatment failure and poor patient survival. The reduced responsiveness of cancer cells is a multifaceted phenomenon that can arise from genetic, epigenetic, and microenvironmental factors. Various mechanisms have been discovered and extensively studied, including drug inactivation, reduced intracellular drug accumulation by reduced uptake or increased efflux, drug target alteration, activation of compensatory pathways for cell survival, regulation of DNA repair and cell death, tumor plasticity, and the regulation from tumor microenvironments (TMEs). To overcome cancer resistance, a variety of strategies have been proposed, which are designed to enhance the effectiveness of cancer treatment or reduce drug resistance. These include identifying biomarkers that can predict drug response and resistance, identifying new targets, developing new targeted drugs, combination therapies targeting multiple signaling pathways, and modulating the TME. The present article focuses on the different mechanisms of drug resistance in cancer and the corresponding tackling approaches with recent updates. Perspectives on polytherapy targeting multiple resistance mechanisms, novel nanoparticle delivery systems, and advanced drug design tools for overcoming resistance are also reviewed.
Collapse
Affiliation(s)
- Zi‐Ning Lei
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Qin Tian
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Qiu‐Xu Teng
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - John N. D. Wurpel
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Leli Zeng
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Yihang Pan
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| |
Collapse
|
9
|
Parveen N, Abourehab MA, Shukla R, Thanikachalam PV, Jain GK, Kesharwani P. Immunoliposomes as an emerging nanocarrier for breast cancer therapy. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2022.111781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
10
|
Moudgil A, Salve R, Gajbhiye V, Chaudhari BP. Challenges and emerging strategies for next generation liposomal based drug delivery: An account of the breast cancer conundrum. Chem Phys Lipids 2023; 250:105258. [PMID: 36375540 DOI: 10.1016/j.chemphyslip.2022.105258] [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: 09/20/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
The global cancer burden is witnessing an upsurge with breast cancer surpassing other cancers worldwide. Furthermore, an escalation in the breast cancer caseload is also expected in the coming years. The conventional therapeutic regimens practiced routinely are associated with many drawbacks to which nanotechnological interventions offer a great advantage. But how eminent could liposomes and their advantages be in superseding these existing therapeutic modalities? A solution is reflected in this review that draws attention to a decade-long journey embarked upon by researchers in this wake. This text is a comprehensive discussion of liposomes, the front runners of the drug delivery systems, and their active and passive targeting approaches for breast cancer management. Active targeting has been studied over the decade by many receptors overexpressed on the breast cancer cells and passive targeting with many drug combinations. The results converge on the fact that the actively targeted formulations exhibit a superior efficacy over their non-targeted counterparts and the all liposomal formulations are efficacious over the free drugs. This undoubtedly underlines the dominion of liposomal formulations over conventional chemotherapy. These investigations have led to the development of different liposomal formulations with active and passive targeting capacities that could be explored in depth. Acknowledging and getting a deeper insight into the liposomal evolution through time also unveiled many imperfections and unchartered territories that can be explored to deliver dexterous liposomal formulations against breast cancer and more in the clinical trial pipeline.
Collapse
Affiliation(s)
- Aliesha Moudgil
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Rajesh Salve
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune 411004, India.
| | - Bhushan P Chaudhari
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pashan, Pune 411008, India.
| |
Collapse
|
11
|
Current advanced drug delivery systems: Challenges and potentialities. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103727] [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]
|
12
|
Pablo-Moreno JAD, Serrano LJ, Revuelta L, Sánchez MJ, Liras A. The Vascular Endothelium and Coagulation: Homeostasis, Disease, and Treatment, with a Focus on the Von Willebrand Factor and Factors VIII and V. Int J Mol Sci 2022; 23:ijms23158283. [PMID: 35955419 PMCID: PMC9425441 DOI: 10.3390/ijms23158283] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/27/2022] Open
Abstract
The vascular endothelium has several important functions, including hemostasis. The homeostasis of hemostasis is based on a fine balance between procoagulant and anticoagulant proteins and between fibrinolytic and antifibrinolytic ones. Coagulopathies are characterized by a mutation-induced alteration of the function of certain coagulation factors or by a disturbed balance between the mechanisms responsible for regulating coagulation. Homeostatic therapies consist in replacement and nonreplacement treatments or in the administration of antifibrinolytic agents. Rebalancing products reestablish hemostasis by inhibiting natural anticoagulant pathways. These agents include monoclonal antibodies, such as concizumab and marstacimab, which target the tissue factor pathway inhibitor; interfering RNA therapies, such as fitusiran, which targets antithrombin III; and protease inhibitors, such as serpinPC, which targets active protein C. In cases of thrombophilia (deficiency of protein C, protein S, or factor V Leiden), treatment may consist in direct oral anticoagulants, replacement therapy (plasma or recombinant ADAMTS13) in cases of a congenital deficiency of ADAMTS13, or immunomodulators (prednisone) if the thrombophilia is autoimmune. Monoclonal-antibody-based anti-vWF immunotherapy (caplacizumab) is used in the context of severe thrombophilia, regardless of the cause of the disorder. In cases of disseminated intravascular coagulation, the treatment of choice consists in administration of antifibrinolytics, all-trans-retinoic acid, and recombinant soluble human thrombomodulin.
Collapse
Affiliation(s)
- Juan A. De Pablo-Moreno
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - Luis Javier Serrano
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
| | - Luis Revuelta
- Department of Physiology, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
| | - María José Sánchez
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía, Pablo de Olavide University, 41013 Sevilla, Spain;
| | - Antonio Liras
- Department of Genetics, Physiology and Microbiology, School of Biology, Complutense University, 28040 Madrid, Spain; (J.A.D.P.-M.); (L.J.S.)
- Correspondence:
| |
Collapse
|
13
|
Development of Irinotecan Liposome Armed with Dual-Target Anti-Epidermal Growth Factor Receptor and Anti-Fibroblast Activation Protein-Specific Antibody for Pancreatic Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14061202. [PMID: 35745775 PMCID: PMC9227843 DOI: 10.3390/pharmaceutics14061202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 01/27/2023] Open
Abstract
Pancreatic cancer is one of the most common causes of death in Taiwan. Previous studies have shown that more than 90% of pancreatic cancer cells presented epidermal growth factor receptor (EGFR) cell marker, and this marker is thought to be important as it is related to activation of cancer cell proliferation, angiogenesis, and cancer progression. Moreover, tumor-associated fibroblasts were involved in tumor proliferation and progression. In this study, we fabricated an anti-EGFR and anti-fibroblast activation protein bispecific antibody-targeted liposomal irinotecan (BS−LipoIRI), which could specifically bind to pancreatic cancer cells and tumor-associated fibroblasts. The drug encapsulation efficiency of BS−LipoIRI was 80.95%, and the drug loading was 8.41%. We proved that both pancreatic cancer cells and fibroblasts could be targeted by BS−LipoIRI, which showed better cellular uptake efficacy compared to LipoIRI. Furthermore, an in vivo mouse tumor test indicated that BS−LipoIRI could inhibit pancreatic cancer growth up to 46.2% compared to phosphate-buffered saline control, suggesting that BS−LipoIRI could be useful in clinical cancer treatment.
Collapse
|
14
|
Recent advances in the development of multifunctional lipid-based nanoparticles for co-delivery, combination treatment strategies, and theranostics in breast and lung cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Synthesis, anticancer activity and docking studies of pyrazoline and pyrimidine derivatives as potential epidermal growth factor receptor (EGFR) inhibitors. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
16
|
A comprehensive review on immuno-nanomedicine for breast cancer therapy: Technical challenges and troubleshooting measures. Int Immunopharmacol 2021; 103:108433. [PMID: 34922248 DOI: 10.1016/j.intimp.2021.108433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022]
Abstract
Nanosized drug carriers have received a major attention in cancer therapeutics and theranostics. The immuno-nanomedicine is a combination of monoclonal antibody (mAb)/mAb-drug-nanoparticles. The immuno-nanomedicine offers a promising strategy to target cancer cells. However, the understating of nanotechnology, cancer biology, immunomedicine, and nanoparticle surface chemistry has provided a better clue to prepare the effective immuno-nanomedicine for cancer therapy. Moreover, the selection of nanoparticles type and its composition is essential for development of efficient drug delivery system (DDS) to target the cancer cell site. Immuno-nanomedicine works in the ligand-receptor binding mechanism through the interaction of mAb conjugated nanoparticles and specific antigen over expressed on target cancer cells. Therefore, the selection of specific receptors in the cancer cell and their ligand is important to prepare the active immuno-nanomedicines. Moreover, the factors such as drug loading, entrapment efficiency, size, shape, and ligand conjugation of a nanocarrier are considered as major factors for a better cancer cell, internalization, drug release, and cancer cell ablation. The target-based over-expression of antigen, mAb is engineered and conjugated with nanoparticles for successful targeting of the cancer cells without causing adverse effects to normal cells. Therefore, this review analyzed the fundamental factors in the immuno-nanomedicine for breast cancer and its technical challenges in the fabrication of the antibody alone/and drug conjugated nanoparticles.
Collapse
|
17
|
Durán-Lobato M, López-Estévez AM, Cordeiro AS, Dacoba TG, Crecente-Campo J, Torres D, Alonso MJ. Nanotechnologies for the delivery of biologicals: Historical perspective and current landscape. Adv Drug Deliv Rev 2021; 176:113899. [PMID: 34314784 DOI: 10.1016/j.addr.2021.113899] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/05/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022]
Abstract
Biological macromolecule-based therapeutics irrupted in the pharmaceutical scene generating a great hope due to their outstanding specificity and potency. However, given their susceptibility to degradation and limited capacity to overcome biological barriers new delivery technologies had to be developed for them to reach their targets. This review aims at analyzing the historical seminal advances that shaped the development of the protein/peptide delivery field, along with the emerging technologies on the lead of the current landscape. Particularly, focus is made on technologies with a potential for transmucosal systemic delivery of protein/peptide drugs, followed by approaches for the delivery of antigens as new vaccination strategies, and formulations of biological drugs in oncology, with special emphasis on mAbs. Finally, a discussion of the key challenges the field is facing, along with an overview of prospective advances are provided.
Collapse
|
18
|
Emerging Approaches to Understanding Microvascular Endothelial Heterogeneity: A Roadmap for Developing Anti-Inflammatory Therapeutics. Int J Mol Sci 2021; 22:ijms22157770. [PMID: 34360536 PMCID: PMC8346165 DOI: 10.3390/ijms22157770] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022] Open
Abstract
The endothelium is the inner layer of all blood vessels and it regulates hemostasis. It also plays an active role in the regulation of the systemic inflammatory response. Systemic inflammatory disease often results in alterations in vascular endothelium barrier function, increased permeability, excessive leukocyte trafficking, and reactive oxygen species production, leading to organ damage. Therapeutics targeting endothelium inflammation are urgently needed, but strong concerns regarding the level of phenotypic heterogeneity of microvascular endothelial cells between different organs and species have been expressed. Microvascular endothelial cell heterogeneity in different organs and organ-specific variations in endothelial cell structure and function are regulated by intrinsic signals that are differentially expressed across organs and species; a result of this is that neutrophil recruitment to discrete organs may be regulated differently. In this review, we will discuss the morphological and functional variations in differently originated microvascular endothelia and discuss how these variances affect systemic function in response to inflammation. We will review emerging in vivo and in vitro models and techniques, including microphysiological devices, proteomics, and RNA sequencing used to study the cellular and molecular heterogeneity of endothelia from different organs. A better understanding of microvascular endothelial cell heterogeneity will provide a roadmap for developing novel therapeutics to target the endothelium.
Collapse
|
19
|
Alves ADCS, Bruinsmann FA, Guterres SS, Pohlmann AR. Organic Nanocarriers for Bevacizumab Delivery: An Overview of Development, Characterization and Applications. Molecules 2021; 26:4127. [PMID: 34299401 PMCID: PMC8305806 DOI: 10.3390/molecules26144127] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 01/08/2023] Open
Abstract
Bevacizumab (BCZ) is a recombinant humanized monoclonal antibody against the vascular endothelial growth factor, which is involved in the angiogenesis process. Pathologic angiogenesis is observed in several diseases including ophthalmic disorders and cancer. The multiple administrations of BCZ can cause adverse effects. In this way, the development of controlled release systems for BCZ delivery can promote the modification of drug pharmacokinetics and, consequently, decrease the dose, toxicity, and cost due to improved efficacy. This review highlights BCZ formulated in organic nanoparticles providing an overview of the physicochemical characterization and in vitro and in vivo biological evaluations. Moreover, the main advantages and limitations of the different approaches are discussed. Despite difficulties in working with antibodies, those nanocarriers provided advantages in BCZ protection against degradation guaranteeing bioactivity maintenance.
Collapse
Affiliation(s)
- Aline de Cristo Soares Alves
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (F.A.B.); (S.S.G.)
| | | | | | - Adriana Raffin Pohlmann
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (F.A.B.); (S.S.G.)
| |
Collapse
|
20
|
Cao J, Zhang M, Wang B, Zhang L, Fang M, Zhou F. Chemoresistance and Metastasis in Breast Cancer Molecular Mechanisms and Novel Clinical Strategies. Front Oncol 2021; 11:658552. [PMID: 34277408 PMCID: PMC8281885 DOI: 10.3389/fonc.2021.658552] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/23/2021] [Indexed: 01/16/2023] Open
Abstract
Breast cancer is the most common malignant tumor in females worldwide. Chemotherapy is the standard breast cancer treatment; however, chemoresistance is often seen in patients with metastatic breast cancer. Owing to high heterogeneity, the mechanisms of breast cancer chemoresistance and metastasis have not been fully investigated. The possible molecular mechanisms of chemoresistance in breast cancer include efflux transporters, signaling pathways, non-coding RNAs, and cancer stem cells. However, to overcome this hurdle, the use of novel clinical strategies such as drug carriers, immunotherapy, and autophagy regulation, are being investigated. The goal of this review is to summarize the current data about the molecular mechanisms of breast cancer chemoresistance and the novel clinical strategies; thus, providing a useful clinical tool to explore optimal treatment for breast cancer.
Collapse
Affiliation(s)
- Jun Cao
- Department of Rare and Head and Neck Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Mengdi Zhang
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Bin Wang
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Long Zhang
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Meiyu Fang
- Department of Rare and Head and Neck Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| |
Collapse
|
21
|
Ortega L, Lobos-González L, Reyna-Jeldes M, Cerda D, De la Fuente-Ortega E, Castro P, Bernal G, Coddou C. The Ω-3 fatty acid docosahexaenoic acid selectively induces apoptosis in tumor-derived cells and suppress tumor growth in gastric cancer. Eur J Pharmacol 2021; 896:173910. [PMID: 33508285 DOI: 10.1016/j.ejphar.2021.173910] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 11/25/2022]
Abstract
Despite current achievements and innovations in cancer treatment, conventional chemotherapy has several limitations, such as unsatisfactory long-term survival, cancer drug resistance and toxicity against non-tumoral cells. In the search for safer therapeutic alternatives, docosahexaenoic acid (DHA) has shown promising effects inhibiting tumor growth without significant side effects in several types of cancer, but in gastric cancer (GC) its effects have not been completely described. In this study, we characterized the effects of DHA in GC using in vivo and in vitro models. Among all of the evaluated Ω-3 and Ω-6 fatty acids, DHA showed the highest antiproliferative potency and selectivity against the GC-derived cell line AGS. 10-100 μM DHA decreased AGS cell viability in a concentration-dependent manner but had no effect on non-tumoral GES-1 cells. To evaluate if the effects of DHA were due to apoptosis induction, cells were stained with Annexin V-PI, observing that 75 and 100 μM DHA increased apoptosis in AGS, but not in GES-1 cells. Additionally, levels of several proapoptotic and antiapoptotic regulators were assessed by qPCR, western blot and activity assays, showing similar results. In order to evaluate DHA efficacy in vivo, xenografts in an immunodeficient mouse model (BALB/cNOD-SCID) were used. In these experiments, DHA treatment for six weeks consistently reduced subcutaneous tumor size, ascitic fluid volume and liver metastasis. In summary, we found that DHA has a selective antiproliferative effect on GC, being this effect driven by apoptosis induction. Our investigation provides promising features for DHA as potential therapeutic agent in GC.
Collapse
Affiliation(s)
- Lorena Ortega
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo, Chile
| | - Lorena Lobos-González
- Centro de Medicina Regenerativa, Facultad de Medicina-Clínica Alemana, Universidad Del Desarrollo, Santiago, Chile; Fundación Ciencia y Vida, Santiago, Chile
| | - Mauricio Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo, Chile; Millennium Nucleus for the Study of Pain (MiNuSPain), Chile
| | - Daniela Cerda
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo, Chile
| | - Erwin De la Fuente-Ortega
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo, Chile
| | - Patricio Castro
- Laboratory of Physiology and Pharmacology for Neural Development, LAND, Facultad de Ciencias Biológicas, Universidad de Concepción, Chile
| | - Giuliano Bernal
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo, Chile
| | - Claudio Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo, Chile; Millennium Nucleus for the Study of Pain (MiNuSPain), Chile.
| |
Collapse
|
22
|
Torres-Martinez Z, Delgado Y, Ferrer-Acosta Y, Suarez-Arroyo IJ, Joaquín-Ovalle FM, Delinois LJ, Griebenow K. Key genes and drug delivery systems to improve the efficiency of chemotherapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:163-191. [PMID: 34142021 PMCID: PMC8208690 DOI: 10.20517/cdr.2020.64] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer cells can develop resistance to anticancer drugs, thereby becoming tolerant to treatment through different mechanisms. The biological mechanisms leading to the generation of anticancer treatment resistance include alterations in transmembrane proteins, DNA damage and repair mechanisms, alterations in target molecules, and genetic responses, among others. The most common anti-cancer drugs reported to develop resistance to cancer cells include cisplatin, doxorubicin, paclitaxel, and fluorouracil. These anticancer drugs have different mechanisms of action, and specific cancer types can be affected by different genes. The development of drug resistance is a cellular response which uses differential gene expression, to enable adaptation and survival of the cell to diverse threatening environmental agents. In this review, we briefly look at the key regulatory genes, their expression, as well as the responses and regulation of cancer cells when exposed to anticancer drugs, along with the incorporation of alternative nanocarriers as treatments to overcome anticancer drug resistance.
Collapse
Affiliation(s)
- Zally Torres-Martinez
- Chemistry Department, University of Puerto Rico- Rio Piedras campus, San Juan, PR 00936, USA
| | - Yamixa Delgado
- Biochemistry & Pharmacology Department, San Juan Bautista School of Medicine, Caguas, PR 00726, USA
| | - Yancy Ferrer-Acosta
- Neuroscience Department, Universidad Central del Caribe, Bayamon, PR 00956, USA
| | | | - Freisa M Joaquín-Ovalle
- Chemistry Department, University of Puerto Rico- Rio Piedras campus, San Juan, PR 00936, USA
| | - Louis J Delinois
- Chemistry Department, University of Puerto Rico- Rio Piedras campus, San Juan, PR 00936, USA
| | - Kai Griebenow
- Chemistry Department, University of Puerto Rico- Rio Piedras campus, San Juan, PR 00936, USA
| |
Collapse
|
23
|
Jurczyk M, Jelonek K, Musiał-Kulik M, Beberok A, Wrześniok D, Kasperczyk J. Single- versus Dual-Targeted Nanoparticles with Folic Acid and Biotin for Anticancer Drug Delivery. Pharmaceutics 2021; 13:326. [PMID: 33802531 PMCID: PMC8001342 DOI: 10.3390/pharmaceutics13030326] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer is one of the major causes of death worldwide and its treatment remains very challenging. The effectiveness of cancer therapy significantly depends upon tumour-specific delivery of the drug. Nanoparticle drug delivery systems have been developed to avoid the side effects of the conventional chemotherapy. However, according to the most recent recommendations, future nanomedicine should be focused mainly on active targeting of nanocarriers based on ligand-receptor recognition, which may show better efficacy than passive targeting in human cancer therapy. Nevertheless, the efficacy of single-ligand nanomedicines is still limited due to the complexity of the tumour microenvironment. Thus, the NPs are improved toward an additional functionality, e.g., pH-sensitivity (advanced single-targeted NPs). Moreover, dual-targeted nanoparticles which contain two different types of targeting agents on the same drug delivery system are developed. The advanced single-targeted NPs and dual-targeted nanocarriers present superior properties related to cell selectivity, cellular uptake and cytotoxicity toward cancer cells than conventional drug, non-targeted systems and single-targeted systems without additional functionality. Folic acid and biotin are used as targeting ligands for cancer chemotherapy, since they are available, inexpensive, nontoxic, nonimmunogenic and easy to modify. These ligands are used in both, single- and dual-targeted systems although the latter are still a novel approach. This review presents the recent achievements in the development of single- or dual-targeted nanoparticles for anticancer drug delivery.
Collapse
Affiliation(s)
- Magdalena Jurczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Katarzyna Jelonek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
| | - Monika Musiał-Kulik
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland; (A.B.); (D.W.)
| | - Janusz Kasperczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, 41-819 Zabrze, Poland; (M.J.); (M.M.-K.); (J.K.)
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland
| |
Collapse
|
24
|
Wang B, Guo C, Liu Y, Han G, Li Y, Zhang Y, Xu H, Chen D. Novel nano-pomegranates based on astragalus polysaccharides for targeting ERα-positive breast cancer and multidrug resistance. Drug Deliv 2021; 27:607-621. [PMID: 32308054 PMCID: PMC7191906 DOI: 10.1080/10717544.2020.1754529] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy is an important method for treating breast cancer. However, multidrug resistance is one of the major challenges in breast cancer chemotherapy. There is an urgent need to develop novel, effective antitumor strategies that will perfect existing therapeutic regimens. In this study, the double-targeted nanocarrier, Quercetin-3'3-dithiodipropionic acid-Astragalus polysaccharides-Folic acid (QDAF), was successfully synthesized and self-assembled into a neoteric nano-targeted delivery strategy, named nano-pomegranates, and which were utilized to effectively inhibit multidrug resistance in estrogen receptor α (ERα)-positive breast tumor. The outstanding abilities of nano-pomegranates to release the drug in a reducing environment was determined by in vitro release assay. The cellular studies in MCF-7 cells were examined that nano-pomegranates have remarkable efficiencies of enhancing cellular uptake, inhibition and necrosis and apoptosis. In vivo antitumor experiments showed that nano-pomegranates have better anti-tumor effects and lower systemic toxicity than free Cur. In conclusion, nano-pomegranates have great potential in anti-breast cancer treatment.
Collapse
Affiliation(s)
- Bingjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China
| | - Chunjing Guo
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China
| | - Yanhui Liu
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| | - Guangting Han
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| | - Yi Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China.,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| | - Yanchun Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, P. R. China
| | - Daquan Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, P.R. China.,State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, P. R. China
| |
Collapse
|
25
|
Florczak A, Deptuch T, Lewandowska A, Penderecka K, Kramer E, Marszalek A, Mackiewicz A, Dams-Kozlowska H. Functionalized silk spheres selectively and effectively deliver a cytotoxic drug to targeted cancer cells in vivo. J Nanobiotechnology 2020; 18:177. [PMID: 33261651 PMCID: PMC7709326 DOI: 10.1186/s12951-020-00734-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/19/2020] [Indexed: 12/31/2022] Open
Abstract
Background Chemotherapy is often a first-line therapeutic approach for the treatment of a wide variety of cancers. Targeted drug delivery systems (DDSs) can potentially resolve the problem of chemotherapeutic drug off-targeting effects. Herein, we examined in vivo models to determine the efficacy of Her2-targeting silk spheres (H2.1MS1) as DDSs for delivering doxorubicin (Dox) to Her2-positive and Her2-negative primary and metastatic mouse breast cancers. Results The specific accumulation of H2.1MS1 spheres was demonstrated at the site of Her2-positive cancer. Dox delivered only by functionalized H2.1MS1 particles selectively inhibited Her2-positive cancer growth in primary and metastatic models. Moreover, the significant effect of the Dox dose and the frequency of treatment administration on the therapeutic efficacy was indicated. Although the control MS1 spheres accumulated in the lungs in Her2-positive metastatic breast cancer, the Dox-loaded MS1 particles did not treat cancer. Histopathological examination revealed no systemic toxicity after multiple administrations and at increased doses of Dox-loaded silk spheres. Although the studies were performed in immunocompetent mice, the H2.1MS1 silk spheres efficiently delivered the drug, which exerted a therapeutic effect. Conclusion Our results indicated that functionalized silk spheres that enable cell-specific recognition, cellular internalization, and drug release represent an efficient strategy for cancer treatment in vivo.![]()
Collapse
Affiliation(s)
- Anna Florczak
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland. .,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.
| | - Tomasz Deptuch
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Anna Lewandowska
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.,Department of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland
| | - Karolina Penderecka
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Elzbieta Kramer
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Andrzej Marszalek
- Department of Tumor Pathology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.,Department of Tumor Pathology and Prophylaxis, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 15 Garbary St, 61-866, Poznan, Poland. .,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-866, Poznan, Poland.
| |
Collapse
|
26
|
Akhter MH, Beg S, Tarique M, Malik A, Afaq S, Choudhry H, Hosawi S. Receptor-based targeting of engineered nanocarrier against solid tumors: Recent progress and challenges ahead. Biochim Biophys Acta Gen Subj 2020; 1865:129777. [PMID: 33130062 DOI: 10.1016/j.bbagen.2020.129777] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Background In past few decades, the research on engineered nanocarriers (NCs) has gained significant attention in cancer therapy due to selective delivery of drug molecules on the diseased cells thereby preventing unwanted uptake into healthy cells to cause toxicity. Scope of review The applicability of enhanced permeability and retention (EPR) effect for the delivery of nanomedicines in cancer therapy has gained limited success due to poor accessibility of the drugs to the target cells where non-specific payload delivery to the off target region lack substantial reward over the conventional therapeutic systems. Major conclusions In spite of the fact, nanomedicines fabricated from the biocompatible nanocarriers have reduced targeting potential for meaningful clinical benefits. However, over expression of receptors on the tumor cells provides opportunity to design functional nanomedicine to bind substantially and deliver therapeutics to the cells or tissues of interest by alleviating the bio-toxicity and unwanted effects. This critique will give insight into the over expressed receptor in various tumor and targeting potential of functional nanomedicine as new therapeutic avenues for effective treatment. General significance This review shortly shed light on EPR-based drug targeting using nanomedicinal strategies, their limitation, and advances in therapeutic targeting to the tumor cells.
Collapse
Affiliation(s)
- Md Habban Akhter
- Department of Pharmaceutics, Faculty of Pharmacy, DIT University, Dehradun, India
| | - Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Mohammed Tarique
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Arshi Malik
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Sarah Afaq
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Biochemistry, Cancer Metabolism & Epigenetic Unit, Faculty of Science, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salman Hosawi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
27
|
Juan A, Cimas FJ, Bravo I, Pandiella A, Ocaña A, Alonso-Moreno C. Antibody Conjugation of Nanoparticles as Therapeutics for Breast Cancer Treatment. Int J Mol Sci 2020; 21:E6018. [PMID: 32825618 PMCID: PMC7504566 DOI: 10.3390/ijms21176018] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer is the most common invasive tumor in women and the second leading cause of cancer-related death. Nanomedicine raises high expectations for millions of patients as it can provide better, more efficient, and affordable healthcare, and it has the potential to develop novel therapeutics for the treatment of solid tumors. In this regard, targeted therapies can be encapsulated into nanocarriers, and these nanovehicles are guided to the tumors through conjugation with antibodies-the so-called antibody-conjugated nanoparticles (ACNPs). ACNPs can preserve the chemical structure of drugs, deliver them in a controlled manner, and reduce toxicity. As certain breast cancer subtypes and indications have limited therapeutic options, this field provides hope for the future treatment of patients with difficult to treat breast cancers. In this review, we discuss the application of ACNPs for the treatment of this disease. Given the fact that ACNPs have shown clinical activity in this clinical setting, special emphasis on the role of the nanovehicles and their translation to the clinic is placed on the revision.
Collapse
Affiliation(s)
- Alberto Juan
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Francisco J. Cimas
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Centro Regional de Investigaciones Biomédicas, Unidad Oncología Traslacional, 02071 Albacete, Spain
| | - Iván Bravo
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer-CSIC, IBSAL- Salamanca and CIBERONC, 37007 Salamanca, Spain;
| | - Alberto Ocaña
- Oncología Traslacional, Unidad de Investigación del Complejo Hospitalario Universitario de Albacete, 02008 Albacete, Spain; (A.J.); (F.J.C.)
- Experimental Therapeutics Unit, Hospital clínico San Carlos, IdISSC and CIBERONC, 28040 Madrid, Spain
| | - Carlos Alonso-Moreno
- Centro Regional de Investigaciones Biomédicas, Unidad NanoCRIB, 02008 Albacete, Spain;
- School of Pharmacy, University of Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
28
|
Shafi S, Khan S, Hoda F, Fayaz F, Singh A, Khan MA, Ali R, Pottoo FH, Tariq S, Najmi AK. Decoding Novel Mechanisms and Emerging Therapeutic Strategies in Breast Cancer Resistance. Curr Drug Metab 2020; 21:199-210. [DOI: 10.2174/1389200221666200303124946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/12/2019] [Accepted: 12/30/2019] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC), an intricate and highly heterogeneous disorder, has presently afflicted 2.09 million females globally. Chemoresistance remains a paramount challenge in the treatment of BC. Owing to its assorted nature, the chemoresistant mechanisms of BC still need intensive research. Accumulating evidence suggests that abnormalities related to the biogenesis of cancer stem cells (CSCs) and microRNAs (miRNAs) are associated with BC progression and chemoresistance. The presently available interventions are inadequate to target chemoresistance, therefore more efficient alternatives are urgently needed to improvise existing therapeutic regimens. A myriad of strategies is being explored, such as immunotherapy, gene therapy, and combination treatment to surmount chemoresistance. Additionally, nanoparticles as chemotherapeutic carriers put forward the options to encapsulate numerous drugs, alone as well as in combination for cancer theranostics. This review summarizes the chemoresistance mechanisms of miRNAs and CSCs as well as the most recently documented therapeutic approaches for the treatment of chemoresistance in BC. By unraveling the underpinning mechanism of BC chemoresistance, researchers could possibly develop more efficient treatment strategies towards BC.
Collapse
Affiliation(s)
- Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Farazul Hoda
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Faizana Fayaz
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Archu Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Sana Tariq
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
29
|
Kumar G, Nandakumar K, Mutalik S, Rao CM. Biologicals to direct nanotherapeutics towards HER2-positive breast cancers. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 27:102197. [PMID: 32275958 DOI: 10.1016/j.nano.2020.102197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 02/17/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022]
Abstract
HER2-positive breast cancer, an aggressive cancer, is treated with combinations of conventional anticancer drugs viz., cytotoxic drugs, nibs, and mAbs. Major limitations associated with this therapy are patient non-compliance due to the adverse drug reactions and rapid development of resistance by the HER2-positive malignant cells. While the former is addressed by the nano-formulations of the anticancer-drugs to some extent, the latter is still at large. This is because the nanocarriers of the anticancer drugs, by and large, lack the target specificity and selectivity. Thus, nowadays, to overcome these problems, various safe and efficacious biological agents are being used to direct the nanotherapeutics towards the HER2-positive breast cancers. The present review describes the potentials of such biological agents.
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Chamallamudi Mallikarjuna Rao
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
30
|
Anaya-Ruiz M, Bandala C, Landeta G, Martínez-Morales P, Zumaquero-Rios JL, Sarracent-Pérez J, Pérez-Santos M. Nanostructured Systems in Advanced Drug Targeting for the Cancer Treatment: Recent Patents. Recent Pat Anticancer Drug Discov 2019; 14:85-94. [PMID: 30381087 DOI: 10.2174/1574892813666181031154146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 09/23/2018] [Accepted: 10/30/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cancer is one of the leading causes of death in the world and it is necessary to develop new strategies for its treatment because most therapies have limited access to many types of tumors, as well as low therapeutic efficacy and high toxicity. OBJECTIVE The present research aims to identify recent patents of drug delivery nanostructured systems that may have application in improving cancer treatment. METHODS Recent patents regarding the drug delivery nanostructured systems for cancer treatment were obtained from the patent databases of the six main patent offices of the world: United States Patent and Trademark Office, European Patent Office, World Intellectual Property Organization, Japan Patent Office, State Intellectual Property Office of China and Korean Intellectual Property Office. RESULTS A total of 1710 patent documents from 1998 to 2017 including "drug delivery nanostructured systems for cancer treatment" were retrieved. The top five countries in patent share were USA, China, South Korea, Canada and Germany. The universities and enterprises of USA had the highest amount of patents followed by institutions from China. CONCLUSION There is a strong tendency for the development of new nanostructured systems for the release of drugs; particularly, in recent years, the development of nanoparticles has focused on nanodiscs, gold nanoparticles and immunoliposomes.
Collapse
Affiliation(s)
- Maricruz Anaya-Ruiz
- Cell Biology Laboratory, Biomedical Research Center of the East, Mexican Institute of Social Security, Metepec, Puebla CP 74360, Mexico
| | - Cindy Bandala
- Department of Neuurosciences, National Institute of Rehabilitation, City of Mexico, Mexico
| | - Gerardo Landeta
- Coordination of Applied Research, University Center for Linkage and Transfer of Technology, Benemérita Autonomous University of Puebla, Puebla CP 72570, Mexico
| | - Patricia Martínez-Morales
- Laboratory of Molecular Biology, Center for Biomedical Research of the East, Mexican Institute of Social Security / CONACYT, Metepec, Puebla CP 74360, Mexico
| | - Jose L Zumaquero-Rios
- Laboratory of Parasitology and Vectors, Faculty of Biology, Benemérita Universidad Autonomous University of Puebla, Puebla CP 72570, Mexico
| | - Jorge Sarracent-Pérez
- Laboratory of Parasitology, Institute of Tropical Medicine "Pedro Kouri ", Havana CP 11400, Cuba
| | - Martín Pérez-Santos
- Technology Marketing Office, University Link Center and Technology Transfer, Benemerita Universidad Autonoma de Puebla, Puebla CP 72570, Mexico
| |
Collapse
|
31
|
Current status of nanomedicine in the chemotherapy of breast cancer. Cancer Chemother Pharmacol 2019; 84:689-706. [DOI: 10.1007/s00280-019-03910-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/25/2019] [Indexed: 12/24/2022]
|
32
|
Li X, Li Q, Jin X, Guo H, Li Y. Long non-coding RNA H19 knockdown inhibits the cell viability and promotes apoptosis of thyroid cancer cells through regulating the PI3K/AKT pathway. Exp Ther Med 2019; 18:1863-1869. [PMID: 31410148 DOI: 10.3892/etm.2019.7720] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/10/2019] [Indexed: 01/02/2023] Open
Abstract
Certain long non-coding (lnc)RNAs have been reported to serve important roles in the genesis and progression of thyroid cancer (TC). Recent studies have demonstrated that the expression of lncRNA H19 is upregulated in TC tissues; however, knowledge of the associated molecular mechanisms is limited. Therefore, the present study aimed to clarify the roles of H19 in TC. The mRNA expression of lncRNA H19 in TC tissues was determined using reverse transcription-quantitative polymerase chain reaction analysis, and the effects of H19 knockdown on cell viability and apoptosis in vitro were assessed using MTT and flow cytometric assays, respectively. Finally, the signaling pathways involved in the effects of H19 were examined. The results indicated that H19 was upregulated in TC tissues. Silencing of H19 inhibited the cell viability and promoted apoptosis of FTC-133 and TPC-1 TC cells, accompanied by an increased expression of B-cell lymphoma 2 (Bcl-2)-associated X protein and caspase 3, and repressed expression of Bcl-2. The results of western blot analysis suggested that the levels of phosphorylated phosphoinositide-3 kinase (PI3K) and phosphorylated AKT were attenuated by H19 silencing. These results suggest that lncRNA H19 exerts an oncogenic function in TC, in part through the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Xiaoyu Li
- Department of Thyroid and Breast Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Qinghuai Li
- Department of Thyroid and Breast Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiao Jin
- Department of Thyroid and Breast Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Hao Guo
- Department of Thyroid and Breast Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yong Li
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
33
|
Zheng Z, Chen M, Xing P, Yan X, Xie B. Increased Expression of Exosomal AGAP2-AS1 (AGAP2 Antisense RNA 1) In Breast Cancer Cells Inhibits Trastuzumab-Induced Cell Cytotoxicity. Med Sci Monit 2019; 25:2211-2220. [PMID: 30910994 PMCID: PMC6446658 DOI: 10.12659/msm.915419] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Trastuzumab therapy is important for patients with HER2-positive breast cancer, but more and more patients have experienced trastuzumab resistance during recent years. Accumulating evidence from recent studies showed that long non-coding RNAs (lncRNAs) play essential roles in chemoresistance of various cancer types, but the precise role of lncRNAs in trastuzumab resistance is unclear. In the present study, we aimed to identify the biofunction of lncRNA APAP2-AS1 in tranastuzumab resistance and to reveal the underlying regulatory mechanism. Material/Methods By culturing HER2-positive SKBR-3 and BT474 cells with transtuzumab-containing medium, we built trastuzumab-resistant cells. Quantitative real-time PCR was used to test the expression of AGAP2-AS1 in the built trastuzumab-resistant cells. Cell viability assay and TUNEL assay were used to test the cell viability and apoptosis in each group. Exosomes were purified from cells cultured in exosomes-depleted FBS and identified by transmission electron microscopy. Results qRT-PCR assay suggested that AGAP2-AS1 was upregulated in the built trastuzumab-resistant cells when compared with parental sensitive cells. Cell viability assay showed that silencing of AGAP2-AS1 enhanced the cytotoxicity induced by trastuzumab treatment. Mechanistically, we revealed that AGAP2-AS1 was secreted outside cells by incorporation into exosomes in an hnRNPA2B1-dependent manner. More importantly, co-culture AGAP2-AS1-containing exosomes with sensitive cells reduced the trastuzumab-induced cell death, and silencing of AGAP2-AS1 from exosomes reversed this effect. In summary, AGAP2-AS1 promotes trastuzumab resistance of breast cancer cells through packaging into exosomes. Conclusions Knockdown of AGAP2-AS1 may be helpful for improving the clinical outcome for HER2+ breast cancer patients and could serve as a therapeutic target.
Collapse
Affiliation(s)
- Zhongqiu Zheng
- Department of Breast and Thyroid Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China (mainland)
| | - Minlong Chen
- Department of Breast and Thyroid Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China (mainland)
| | - Ping Xing
- Department of Breast and Thyroid Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China (mainland)
| | - Xingqiang Yan
- Department of Breast and Thyroid Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China (mainland)
| | - Bojian Xie
- Department of Breast and Thyroid Surgery, Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang, China (mainland)
| |
Collapse
|
34
|
Chemoresistance mechanisms of breast cancer and their countermeasures. Biomed Pharmacother 2019; 114:108800. [PMID: 30921705 DOI: 10.1016/j.biopha.2019.108800] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/13/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Chemoresistance is one of the major challenges for the breast cancer treatment. Owing to its heterogeneous nature, the chemoresistance mechanisms of breast cancer are complicated, and not been fully elucidated. The existing treatments fall short of offering adequate solution to drug resistance, so more effective approaches are desperately needed to improve existing therapeutic regimens. To overcome this hurdle, a number of strategies are being investigated, such as novel agents or drug carriers and combination treatment. In addition, some new therapeutics including gene therapy and immunotherapy may be promising for dealing with the resistance. In this article, we review the mechanisms of chemoresistance in breast cancer. Furthermore, the potential therapeutic methods to overcome the resistance were discussed.
Collapse
|
35
|
Zhang J, Song J, Liang X, Yin Y, Zuo T, Chen D, Shen Q. Hyaluronic acid-modified cationic nanoparticles overcome enzyme CYP1B1-mediated breast cancer multidrug resistance. Nanomedicine (Lond) 2019; 14:447-464. [PMID: 30694105 DOI: 10.2217/nnm-2018-0244] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Aim: Enzyme CYP1B1 (CYP1B1) is usually overexpressed in multidrug resistance (MDR) breast cancer cells, which could metabolically inactivate docetaxel (DTX). Materials & methods: The cationic core–shell nanoparticles (hyaluronic acid/polyethyleneimine nanoparticles [HA/PEI NPs]) modified with hyaluronic acid (HA) were developed and coloaded with DTX and α-napthtoflavone (ANF, a CYP1B1 inhibitor) to overcome MDR in breast cancer induced by CYP1B1. Physicochemical characterization, MDR reversing effect in vitro and pharmacokinetics in vivo of HA/PEI NPs were evaluated. Results: The HA/PEI NPs exhibited spherical morphology with size of (193.6 ± 3.1) nm. The HA/PEI NPs could reverse MDR effectively by downregulating the expression of CYP1B1. The HA/PEI NPs improved the bioavailability of DTX. Conclusion: The HA/PEI NPs might be a promising strategy to overcome CYP1B1-mediated breast cancer MDR.
Collapse
Affiliation(s)
- Jun Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Jia Song
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Xiao Liang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Yunzhi Yin
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Tiantian Zuo
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qi Shen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
36
|
Cheng YA, Chen IJ, Su YC, Cheng KW, Lu YC, Lin WW, Hsieh YC, Kao CH, Chen FM, Roffler SR, Cheng TL. Enhanced drug internalization and therapeutic efficacy of PEGylated nanoparticles by one-step formulation with anti-mPEG bispecific antibody in intrinsic drug-resistant breast cancer. Biomater Sci 2019; 7:3404-3417. [DOI: 10.1039/c9bm00323a] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
One-step formulation of BsAb with PLD is a simple method to enhance tumor specificity, internalization and the anti-cancer activity.
Collapse
|
37
|
Qu N, Shi D, Shang M, Duan S, Guo L, Ning S, Li J. Breast Cancer Cell Line Phenotype Affects Sonoporation Efficiency Under Optimal Ultrasound Microbubble Conditions. Med Sci Monit 2018; 24:9054-9062. [PMID: 30546004 PMCID: PMC6302661 DOI: 10.12659/msm.910790] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Ultrasound/microbubble (USMB)-mediated sonoporation is a new strategy with minimal procedural invasiveness for targeted and site-specific drug delivery to tumors. The purpose of this study was to explore the effect of different breast cancer cell lines on sonoporation efficiency, and then to identify an optimal combination of USMB parameters to maximize the sonoporation efficiency for each tumor cell line. Material/Methods Three drug-sensitive breast cell lines – MCF-7, MDA-MB-231, and MDA-MB-468 – and 1 multidrug resistance (MDR) cell line – MCF-7/ADR – were chosen. An orthogonal array experimental design approach based on 3 levels of 3 parameters (A: microbubble concentration, 10%, 20%, and 30%, B: sound intensity, 0.5, 1.0, and 1.5 W/cm2, C: irradiation time, 30, 60, and 90 s) was employed to optimize the sonoporation efficiency. Results The optimal USMB parameter combinations for different cell lines were diverse. Under optimal parameter combinations, the maximum sonoporation efficiency differences between different breast tumor cell lines were statistically significant (MDA-MB-231: 46.70±5.79%, MDA-MB-468: 53.44±5.69%, MCF-7: 59.88±5.53%, MCF-7/ADR: 65.39±4.01%, P<0.05), so were between drug-sensitive cell line and MDR cell line (MCF-7: 59.88±5.53%, MCF-7/ADR: 65.39±4.01%, p=0.026). Conclusions Different breast tumor cell lines have their own optimal sonoporation. Drug-resistant MCF-7/ADR cells had higher sonoporation efficiency than drug-sensitive MCF-7 cells. The molecular subtype of tumors should be considered when sonoporation is applied, and optimal parameter combination may have the potential to improve drug-delivery efficiency by increasing the sonoporation efficiency.
Collapse
Affiliation(s)
- Nina Qu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland).,Department of Ultrasound, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China (mainland)
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Sujuan Duan
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Song Ning
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
38
|
Lei Y, Guo W, Chen B, Chen L, Gong J, Li W. Tumor‑released lncRNA H19 promotes gefitinib resistance via packaging into exosomes in non‑small cell lung cancer. Oncol Rep 2018; 40:3438-3446. [PMID: 30542738 PMCID: PMC6196604 DOI: 10.3892/or.2018.6762] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/19/2018] [Indexed: 02/05/2023] Open
Abstract
Currently, resistance to tyrosine kinase inhibitors, such as gefitinib, has become one major obstacle for improving the clinical outcome of patients with metastatic and advanced-stage non-small cell lung cancer (NSCLC). While cell behavior can be modulated by long non-coding RNAs (lncRNAs), the contributions of lncRNAs within extracellular vesicles (exosomes) are largely unknown. To this end, the involvement and regulatory functions of lncRNA H19 wrapped by exosomes during formation of gefitinib resistance in human NSCLC were investigated. Gefitinib-resistant cell lines were built by continuously grafting HCC827 and HCC4006 cells into gefitinib-contained culture medium. RT-qPCR assays indicated that H19 was increased in gefitinib-resistant cells when compared to sensitive parent cells. Functional experiments revealed that silencing of H19 potently promoted gefitinib-induced cell cytotoxicity. H19 was secreted by packaging into exosomes and this packaging process was specifically mediated by hnRNPA2B1. H19 wrapped in exosomes could be transferred to non-resistant cells, thus inducing gefitinib resistance. Moreover, treatment-sensitive cells with exosomes highly-expressing H19 induced gefitinib resistance, while knockdown of H19 abrogated this effect. In conclusion, H19 promoted gefitinib resistance of NSCLC cells by packaging into exosomes. Therefore, exosomal H19 may be a promising therapeutic target for EGFR+ NSCLC patients.
Collapse
Affiliation(s)
- Yi Lei
- International Medical Center/Department of General Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wang Guo
- International Medical Center/Department of General Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Bowang Chen
- International Medical Center/Department of General Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lu Chen
- International Medical Center/Department of General Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jiaxin Gong
- International Medical Center/Department of General Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Weimin Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
39
|
Soe ZC, Thapa RK, Ou W, Gautam M, Nguyen HT, Jin SG, Ku SK, Oh KT, Choi HG, Yong CS, Kim JO. Folate receptor-mediated celastrol and irinotecan combination delivery using liposomes for effective chemotherapy. Colloids Surf B Biointerfaces 2018; 170:718-728. [PMID: 30005409 DOI: 10.1016/j.colsurfb.2018.07.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/17/2018] [Accepted: 07/06/2018] [Indexed: 01/08/2023]
Abstract
Drug targeting using functionalized nanoparticles provides a new standard in anticancer therapy. Liposomes, safe and effective drug delivery carriers, can incorporate both hydrophilic and hydrophobic drugs for combination chemotherapy treatment of cancers. The objectives of the current study were to synthesize and test the effectiveness of a nanotechnology-based strategy utilizing folic acid (FA)-conjugated liposomes that incorporate both celastrol (Cs) and irinotecan (Ir) for targeted breast cancer therapy. Our results revealed the successful preparation of Cs and Ir-loaded folate-targeted liposomes (Lipo/Cs/Ir-FA) with a small particle size (∼190 nm) and polydispersity index (∼0.10). The formulation exhibited higher drug release profiles for both Ir and Cs at pH 5.0 compared to those at physiological pH, favoring cancer cell-targeted release. Furthermore, in vitro cell studies showed high uptake and enhanced apoptosis in folate receptor-positive breast cancer cells (MCF-7 and MDA-MB-231), but not in folate receptor-negative lung cancer cells (A549). Moreover, an in vivo study in a mouse tumor model using MDA-MB-231 xenografts supported effective drug delivery behavior of the folate-conjugated liposomes by selective targeting of tumor tissue and minimizing systemic adverse effects. Therefore, our formulation could provide an effective therapy for targeted cancer treatment.
Collapse
Affiliation(s)
- Zar Chi Soe
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, Republic of Korea
| | - Raj Kumar Thapa
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, Republic of Korea
| | - Wenquan Ou
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, Republic of Korea
| | - Milan Gautam
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, Republic of Korea
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, Republic of Korea
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Dongnam-gu, Cheonan, 31116, Republic of Korea
| | - Sae Kwang Ku
- College of Korean Medicine, Daegu Haany University, Gyeongsan, 712-715, Republic of Korea
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 221 Heuksuk-dong Dongjak-gu, Seoul, 156-756, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, 426-791, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-dong, Gyeongsan, 712-749, Republic of Korea.
| |
Collapse
|
40
|
Rusz O, Papp O, Vízkeleti L, Molnár BÁ, Bende KC, Lotz G, Ács B, Kahán Z, Székely T, Báthori Á, Szundi C, Kulka J, Szállási Z, Tőkés AM. LAPTM4B gene copy number gain is associated with inferior response to anthracycline-based chemotherapy in hormone receptor negative breast carcinomas. Cancer Chemother Pharmacol 2018; 82:139-147. [PMID: 29770955 DOI: 10.1007/s00280-018-3602-z] [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: 02/20/2018] [Accepted: 05/12/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE To determine the associations between lysosomal-associated transmembrane protein 4b (LAPTM4B) gene copy number and response to different chemotherapy regimens in hormone receptor negative (HR-) primary breast carcinomas. PATIENTS AND METHODS Two cohorts were analyzed: (1) 69 core biopsies from HR-breast carcinomas treated with neoadjuvant chemotherapy (anthracycline based in 72.5% of patients and non-anthracycline based in 27.5% of patients). (2) Tissue microarray (TMA) of 74 HR-breast carcinomas treated with adjuvant therapy (77.0% of the patients received anthracycline, 17.6% of the patients non-anthracycline-based therapy, and in 5.4% of the cases, no treatment data are available). Interphase FISH technique was applied on pretreatment core biopsies (cohort I) and on TMAs (cohort II) using custom-made dual-labelled FISH probes (LAPTM4B/CEN8q FISH probe Abnova Corp.). RESULTS In the neoadjuvant cohort in the anthracycline-treated group, we observed a significant difference (p = 0.029) of average LAPTM4B copy number between the non-responder and pathological complete responder groups (4.1 ± 1.1 vs. 2.6 ± 0.1). In the adjuvant setting, the anthracycline-treated group of metastatic breast carcinomas was characterized by higher LAPTM4B copy number comparing to the non-metastatic ones (p = 0.046). In contrast, in the non-anthracycline-treated group of patients, we did not find any LAPTM4B gene copy number differences between responder vs. non-responder groups or between metastatic vs. non-metastatic groups. CONCLUSION Our results confirm the possible role of the LAPTM4B gene in anthracycline resistance in HR- breast cancer. Analyzing LAPTM4B copy number pattern may support future treatment decision.
Collapse
Affiliation(s)
- Orsolya Rusz
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, 6720, Hungary
| | - Orsolya Papp
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Laura Vízkeleti
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary.,MTA-SE-NAP B Brain Metastasis Research Group, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Béla Ákos Molnár
- 1st Department of Surgery, Semmelweis University, Üllői út 78, Budapest, 1082, Hungary
| | - Kristóf Csaba Bende
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Gábor Lotz
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Balázs Ács
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, Szeged, 6720, Hungary
| | - Tamás Székely
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Ágnes Báthori
- Department of Pathology, University of Szeged, Állomás u. 2, Szeged, 6725, Hungary
| | - Csilla Szundi
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Janina Kulka
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary
| | - Zoltán Szállási
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary.,MTA-SE-NAP B Brain Metastasis Research Group, 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.,Department of Bio and Health Informatics, Technical University of Denmark, Kemitorvet 208, 2800, Lyngby, Denmark.,Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Harvard University, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Anna-Mária Tőkés
- 2nd Department of Pathology, Semmelweis University, Üllői út 93, Budapest, 1091, Hungary.
| |
Collapse
|
41
|
Abstract
The phenomenal advances in pharmaceutical sciences over the last few decades have led to the development of new therapeutics like peptides, proteins, RNAs, DNAs and highly potent small molecules. Fruitful applications of these therapeutics have been challenged by several anatomical and physiological barriers that limit adequate drug disposition at the site-of-action and by off-target drug distribution to undesired tissues, which together result in the reduced effectiveness and increased side effects of therapeutic agents. As such, the development of drug delivery and targeting systems has been recognised as a cornerstone for future drug development. Research in pharmaceutical sciences is now devoted to tackling delivery challenges through engineering delivery systems that move beyond conventional dosage forms and regimens into state-of-the-art targeted drug delivery tailored toward specific therapeutic needs. Modern drug delivery systems comprise passive and active targeting approaches. While passive targeting relies on the natural course of distribution of drugs or drug carriers in the body, as governed by their physicochemical properties, active targeting often exploits targeting moieties that home preferentially into target tissues. Here, we provide an overview of theories of and approaches to passive and active drug delivery. As the design of drug delivery is dependent on the unique structure of target tissues and organs, we present our discussion in an organ-specific manner with the aim to inspire the development of new strategies for curing disease with high accuracy and efficiency.
Collapse
Affiliation(s)
- Mohammad Alsaggar
- a Department of Pharmaceutical Technology, College of Pharmacy , Jordon University of Science and Technology , Irbid , Jordan
| | - Dexi Liu
- b Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy , University of Georgia , Athens , GA , USA
| |
Collapse
|
42
|
Li Y, Xiao BX, Dong J, Liu Y, Gao S, Pang J, Sun Z. Near-infrared light-responsive nanoparticles for improved anticancer efficacy through synergistic chemo-photothermal therapy. Pharm Dev Technol 2017; 23:116-124. [PMID: 29160121 DOI: 10.1080/10837450.2017.1402934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Combined treatment is more effective than single treatment against most forms of cancer. The synergistic chemo-thermotherapy mediated by nanoparticles has superior advantages of lesser adverse effects as a promising cancer therapy modality. In this study, we report a theranostic carrier system co-encapsulating Doxorubicin (DOX) and Indocyanine green (ICG) into the D-α-Tocopheryl polyethylene glycol 1000 succinate (TPGS). Full physicochemical characterization studies of the DOX/ICG-loaded TPGS nanoparticles (TNPs) are performed. TNPs have a mean size around 60 nm with superior photostability, and entrapment efficiency of drugs in TNPs was 75.0% for ICG and 68.3% for DOX. TNPs also exhibit a longer sustained release with around 63% of the entrapped drug in 24 h. In vitro studies, TNPs could effectively enhance cellular uptake of DOX and ICG, which permitted high therapeutic efficacy against cancer cells. Further, we investigate antitumor efficacy of TNPs along with its impact on major organs in vivo, TNPs also exhibit a complete inhibition of tumor growth and minimal side effects after irradiation. Collectively, these results suggest that near-infrared light-responsive TNPs can further enhance antitumor effects by synergistic chemo-photothermal therapy.
Collapse
Affiliation(s)
- Yanli Li
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Bing Xin Xiao
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Jie Dong
- b Department of College of Pharmaceutical Science , Xuzhou Medical University , Xuzhou , PR China
| | - Yun Liu
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Shan Gao
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Jie Pang
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Zengxian Sun
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| |
Collapse
|
43
|
Wu D, Si M, Xue HY, Wong HL. Nanomedicine applications in the treatment of breast cancer: current state of the art. Int J Nanomedicine 2017; 12:5879-5892. [PMID: 28860754 PMCID: PMC5566389 DOI: 10.2147/ijn.s123437] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the most common malignant disease in women worldwide, but the current drug therapy is far from optimal as indicated by the high death rate of breast cancer patients. Nanomedicine is a promising alternative for breast cancer treatment. Nanomedicine products such as Doxil® and Abraxane® have already been extensively used for breast cancer adjuvant therapy with favorable clinical outcomes. However, these products were originally designed for generic anticancer purpose and not specifically for breast cancer treatment. With better understanding of the molecular biology of breast cancer, a number of novel promising nanotherapeutic strategies and devices have been developed in recent years. In this review, we will first give an overview of the current breast cancer treatment and the updated status of nanomedicine use in clinical setting, then discuss the latest important trends in designing breast cancer nanomedicine, including passive and active cancer cell targeting, breast cancer stem cell targeting, tumor microenvironment-based nanotherapy and combination nanotherapy of drug-resistant breast cancer. Researchers may get insight from these strategies to design and develop nanomedicine that is more tailored for breast cancer to achieve further improvements in cancer specificity, antitumorigenic effect, antimetastasis effect and drug resistance reversal effect.
Collapse
Affiliation(s)
- Di Wu
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Mengjie Si
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Hui-Yi Xue
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Ho-Lun Wong
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| |
Collapse
|