1
|
Parkin HC, Shopperly LK, Perez MR, Willerth SM, Manners I. Uniform block copolymer nanofibers for the delivery of paclitaxel in 2D and 3D glioblastoma tumor models. Biomater Sci 2024; 12:5283-5294. [PMID: 39246052 DOI: 10.1039/d4bm00480a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Cancer treatment has transformed in recent years, with the introduction of immunotherapy providing substantial improvements in prognoses for certain cancers. However, traditional small molecule chemotherapeutics remain the major frontline of defence, and improving their delivery to solid tumors is of utmost importance for improving potency and reducing side effects. Here, length-controlled one-dimensional seed nanofibers (ca. 25 nm, ĐL = 1.05) were generated from poly(fluorenetrimethylenecarbonate)-block-poly(dimethylaminoethylmethacrylate) via living crystallization-driven self-assembly. Paclitaxel, with an encapsulation content ranging from 1 to 100 wt%, was loaded onto the preformed nanoparticles by solvent addition and evaporation. Drug loading was quantified by dynamic light scattering and transmission electron microscopy. Drug-loaded vectors were then incubated with U87 MG glioblastoma cells in a 2D cell assay for up to 72 h, and their anticancer properties were determined. It was observed that seed nanofibers loaded with 20 wt% paclitaxel were the most advantageous combination (IC50 = 0.48 μg mL-1), while pure seed nanofibers with no loaded drug displayed much lower cytotoxicity (IC50 = 11.52 μg mL-1). The IC50 of the loaded seed nanofibers rivaled that of the commercially approved Abraxane® (IC50 = 0.46 μg mL-1). 3D tumor spheroids were then cultured and subjected to the same stresses. Live/dead cell staining revealed that once more, seed nanofibers with 20 wt% paclitaxel, Abraxane®, and paclitaxel all exhibited similar levels of potency (55% viability), whereas control samples exhibited much higher cell viability (70%) after 3 days. These results demonstrate that nanofibers contain great potential as biocompatible drug delivery vehicles for cancer treatment as they exert a similar anticancer effect to the commercially available Abraxane®.
Collapse
Affiliation(s)
- Hayley C Parkin
- Department of Chemistry, University of Victoria, Victoria, BC V8 W 3 V6, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada.
| | - Lennard K Shopperly
- Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, BC V8 W 3 V6, Canada
| | - Milena R Perez
- Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, BC V8 W 3 V6, Canada
| | - Stephanie M Willerth
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada.
- Department of Mechanical Engineering and Division of Medical Sciences, University of Victoria, Victoria, BC V8 W 3 V6, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Ian Manners
- Department of Chemistry, University of Victoria, Victoria, BC V8 W 3 V6, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, 3800 Finnerty Rd, Victoria, BC, V8P 5C2, Canada.
| |
Collapse
|
2
|
Chao CJ, Zhang E, Zhao Z. Engineering cells for precision drug delivery: New advances, clinical translation, and emerging strategies. Adv Drug Deliv Rev 2023; 197:114840. [PMID: 37088403 DOI: 10.1016/j.addr.2023.114840] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Cells have emerged as a promising new form of drug delivery carriers owing to their distinguished advantages such as naturally bypassing immune recognition, intrinsic capability to navigate biological barriers, and access to hard-to-reach tissues via onboarding sensing and active motility. Over the past two decades, a large body of work has focused on understanding the ability of cell carriers to breach biological barriers and to modulate drug pharmacokinetics and pharmacodynamics. These efforts have led to the engineering of various cells for tissue-specific drug delivery. Despite exciting advances, clinical translation of cell-based drug carriers demands a thorough understanding of the pressing challenges and potential strategies to overcome them. Here, we summarize recent advances and new concepts in cell-based drug carriers and their clinical translation. We also discuss key considerations and emerging strategies to engineering the next-generation cell-based delivery technologies for more precise, targeted drug delivery.
Collapse
Affiliation(s)
- Chih-Jia Chao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612.
| |
Collapse
|
3
|
Liu T, Gao C, Gu D, Tang H. Cell-based carrier for targeted hitchhiking delivery. Drug Deliv Transl Res 2022; 12:2634-2648. [PMID: 35499717 DOI: 10.1007/s13346-022-01149-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Drug delivery systems aim at improving drug transport efficiency and therapeutic efficacy by rational design, and current research on conventional delivery systems brings new developments for disease treatment. Recently, studies on cell-based drug delivery systems are rapidly emerging, which shows great advantages in comparison to conventional drug delivery system. The system uses cells as carriers to delivery conventional drugs or nanomedicines and shows good biocompatibility and enhanced targeting efficiency, beneficial from self component and its physiological function. The construction methodology of cell-based carrier determines the effect on the physiological functions of transporting cell and affects its clinical application. There are different strategies to prepare cell-based carrier, such as direct internalization or surface conjugation of drugs or drug loaded materials. Thus, it is necessary to fully understand the advantages and disadvantages of different strategies for constructing cell-based carrier and then to seek the appropriate construction methodology for achieving better therapeutic results based on disease characterization. We here summarize the application of different types of cell-based carriers reported in recent years and further discuss their applications in disease therapy and the dilemmas faced in clinical translation. We hope that this summary can accelerate the process of clinical translation by promoting the technology development of cell-based carrier.
Collapse
Affiliation(s)
- Tonggong Liu
- Department of Preventive Medicine, School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, China.,Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China
| | - Cheng Gao
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.,Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, 518035, China.
| | - Huanwen Tang
- Department of Preventive Medicine, School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
4
|
Jahromi LP, Shahbazi M, Maleki A, Azadi A, Santos HA. Chemically Engineered Immune Cell-Derived Microrobots and Biomimetic Nanoparticles: Emerging Biodiagnostic and Therapeutic Tools. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002499. [PMID: 33898169 PMCID: PMC8061401 DOI: 10.1002/advs.202002499] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/26/2020] [Indexed: 05/16/2023]
Abstract
Over the past decades, considerable attention has been dedicated to the exploitation of diverse immune cells as therapeutic and/or diagnostic cell-based microrobots for hard-to-treat disorders. To date, a plethora of therapeutics based on alive immune cells, surface-engineered immune cells, immunocytes' cell membranes, leukocyte-derived extracellular vesicles or exosomes, and artificial immune cells have been investigated and a few have been introduced into the market. These systems take advantage of the unique characteristics and functions of immune cells, including their presence in circulating blood and various tissues, complex crosstalk properties, high affinity to different self and foreign markers, unique potential of their on-demand navigation and activity, production of a variety of chemokines/cytokines, as well as being cytotoxic in particular conditions. Here, the latest progress in the development of engineered therapeutics and diagnostics inspired by immune cells to ameliorate cancer, inflammatory conditions, autoimmune diseases, neurodegenerative disorders, cardiovascular complications, and infectious diseases is reviewed, and finally, the perspective for their clinical application is delineated.
Collapse
Affiliation(s)
- Leila Pourtalebi Jahromi
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShiraz71468‐64685Iran
- Present address:
Helmholtz Institute for Pharmaceutical Research SaarlandHelmholtz Centre for Infection ResearchBiogenic Nanotherapeutics GroupCampus E8.1Saarbrücken66123Germany
| | - Mohammad‐Ali Shahbazi
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC)Zanjan University of Medical SciencesZanjan45139‐56184Iran
| | - Aziz Maleki
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC)Zanjan University of Medical SciencesZanjan45139‐56184Iran
| | - Amir Azadi
- Pharmaceutical Sciences Research CenterShiraz University of Medical SciencesShiraz71468‐64685Iran
- Department of PharmaceuticsSchool of PharmacyShiraz University of Medical SciencesShiraz71468‐64685Iran
| | - Hélder A. Santos
- Drug Research ProgramDivision of Pharmaceutical Chemistry and TechnologyFaculty of PharmacyUniversity of HelsinkiHelsinkiFI‐00014Finland
- Helsinki Institute of Life Science (HiLIFE)University of HelsinkiHelsinkiFI‐00014Finland
| |
Collapse
|
5
|
Zhou C, Cheng H, Qin W, Zhang Y, Xiong H, Yang J, Huang H, Wang Y, Chen XZ, Tang J. Pygopus2 inhibits the efficacy of paclitaxel-induced apoptosis and induces multidrug resistance in human glioma cells. Oncotarget 2018; 8:27915-27928. [PMID: 28427190 PMCID: PMC5438618 DOI: 10.18632/oncotarget.15843] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/20/2017] [Indexed: 12/19/2022] Open
Abstract
Anti-microtubule drugs, such as paclitaxel (PTX), are extensively used for the treatment of numerous cancers. However, growing evidence has shown that PTX resistance, either intrinsic or acquired, frequently occurs in patients and results in the failure of treatment, contributing to the high cancer mortality rate. Therefore, it is necessary to identify the genes or pathways involved in anti-microtubule drug resistance for future successful treatment of cancers. Pygopus2 (Pygo2), which contains a Zn-coordinated plant homeodomain (PHD) finger domain, is critical for β-catenin-dependent transcriptional switches in normal and malignant tissues and is over-expressed in various cancers, including human brain glioma. In this study, we report that over-expression of Pygo2 inhibited the efficacy of PTX and contributed to cell multidrug resistance in two different ways. First, over-expression of Pygo2 inhibited the PTX-induced phosphorylation of B-cell lymphoma 2 (Bcl-2), suppressing the proteolytic cleavage of procaspase-8/9 and further inhibiting the activation of caspase-3, which also inhibits the activation of the JNK/SAPK pathway, ultimately inhibiting cell apoptosis. Second, over-expression of Pygo2 facilitated the expression of P-glycoprotein, which acts as a drug efflux pump, by promoting the transcription of Multi-drug resistance 1 (MDR1) at the MDR1 promoter loci, resulting in acceleration of the efflux of PTX.
Collapse
Affiliation(s)
- Cefan Zhou
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering, Hubei University of Technology, Wuhan, 430068, China.,The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Hongxia Cheng
- Department of Chemical and Pharmaceutical Engineering, Wuhan Huaxia University of Technology, 430223, China
| | - Wenying Qin
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering, Hubei University of Technology, Wuhan, 430068, China
| | - Yi Zhang
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering, Hubei University of Technology, Wuhan, 430068, China
| | - Hui Xiong
- XiLi People's Hospital, Shenzhen, Guangdong, 518055, China
| | - Jing Yang
- Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Huang Huang
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering, Hubei University of Technology, Wuhan, 430068, China
| | - Yefu Wang
- The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xing-Zhen Chen
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering, Hubei University of Technology, Wuhan, 430068, China.,Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jingfeng Tang
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), College of Bioengineering, Hubei University of Technology, Wuhan, 430068, China
| |
Collapse
|
6
|
Zheng H, Yang B, Xu D, Wang W, Tan J, Sun L, Li Q, Sun L, Xia X. Induction of specific T helper-9 cells to inhibit glioma cell growth. Oncotarget 2018; 8:4864-4874. [PMID: 28002799 PMCID: PMC5354876 DOI: 10.18632/oncotarget.13981] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
The effects of Staphylococcal enterotoxin B (SEB) on regulation of immune response have been recognized; whether SEB can enhance the effects of immunotherapy on glioma remains to be investigated. This study tests a hypothesis that administration with SEB enhances the effects of specific immunotherapy on glioma growth in mice. In this study, a glioma-bearing mouse model was developed by adoptive transfer with GL261 cells (a mouse glioma cell line). The mice were treated with the GL261 cell extracts (used as an Ag) with or without administration of SEB. We observed that treating glioma-bearing mice with the glioma Ag and SEB induced glioma-specific Th9 cells in both glioma tissue and the spleen. Treating CD4+ CD25− T cells with SEB increased p300 phosphorylation, histone H3K4 acetylation at the interleukin (IL)-9 promoter locus, and increased the IL-9 transcriptional factor binding to the IL-9 promoter. Treating CD4+ CD25− T cells with both SEB and glioma Ag induced glioma-specific Th9 cells. The glioma-specific Th9 cells induced glioma cell apoptosis in the culture. Treating the glioma-bearing mice with SEB and glioma Ag significantly inhibited the glioma growth. In conclusion, SEB plus glioma Ag immunotherapy inhibits the experimental glioma growth, which may be a novel therapeutic remedy for the treatment of glioma.
Collapse
Affiliation(s)
- Haiyan Zheng
- Department of Neurosurgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
| | - Baohua Yang
- Department of Neurosurgery, Guilin Medical University, Affiliated Hospital, Guilin, 541001, China
| | - Dedong Xu
- Department of Neurosurgery, Hainan General Hospital, Haikou, 570311, China
| | - Wenbo Wang
- Department of Neurosurgery, Guilin Medical University, Affiliated Hospital, Guilin, 541001, China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541001, China
| | - Liyuan Sun
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541001, China
| | - Qinghua Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541001, China
| | - Li Sun
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541001, China
| | - Xuewei Xia
- Department of Neurosurgery, Guilin Medical University, Affiliated Hospital, Guilin, 541001, China.,Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541001, China
| |
Collapse
|
7
|
Bhavsar C, Momin M, Khan T, Omri A. Targeting tumor microenvironment to curb chemoresistance via novel drug delivery strategies. Expert Opin Drug Deliv 2018; 15:641-663. [PMID: 29301448 DOI: 10.1080/17425247.2018.1424825] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Tumor is a heterogeneous mass of malignant cells co-existing with non-malignant cells. This co-existence evolves from the initial developmental stages of the tumor and is one of the hallmarks of cancer providing a protumorigenic niche known as tumor microenvironment (TME). Proliferation, invasiveness, metastatic potential and maintenance of stemness through cross-talk between tumors and its stroma forms the basis of TME. AREAS COVERED The article highlights the developmental phases of a tumor from dysplasia to the formation of clinically detectable tumors. The authors discuss the mechanistic stages involved in the formation of TME and its contribution in tumor outgrowth and chemoresistance. The authors have reviewed various approaches for targeting TME and its hallmarks along with their advantages and pitfalls. The authors also highlight cancer stem cells (CSCs) that are resistant to chemotherapeutics and thus a primary reason for tumor recurrence thereby, posing a challenge for the oncologists. EXPERT OPINION Recent understanding of the cellular and molecular mechanisms involved in acquired chemoresistance has enabled scientists to target the tumor niche and TME and modulate and/or disrupt this communication leading to the transformation from a tumor-supportive niche environment to a tumor-non-supporting environment and give synergistic results towards an effective management of cancer.
Collapse
Affiliation(s)
- Chintan Bhavsar
- a Department of Pharmaceutics, SVKMs Dr. Bhanuben Nanavati College of Pharmacy , University of Mumbai , Mumbai , India
| | - Munira Momin
- a Department of Pharmaceutics, SVKMs Dr. Bhanuben Nanavati College of Pharmacy , University of Mumbai , Mumbai , India
| | - Tabassum Khan
- b Department of Quality Assurance and Pharmaceutical Chemistry, SVKMs Dr. Bhanuben Nanavati College of Pharmacy , University of Mumbai , Mumbai , India
| | - Abdelwahab Omri
- c The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry , Laurentian University , Sudbury , ON , Canada
| |
Collapse
|
8
|
Li Z, Liu P, Zhang H, Zhao S, Jin Z, Li R, Guo Y, Wang X. Role of GABA B receptors and p38MAPK/NF-κB pathway in paclitaxel-induced apoptosis of hippocampal neurons. PHARMACEUTICAL BIOLOGY 2017; 55:2188-2195. [PMID: 29115173 PMCID: PMC6130610 DOI: 10.1080/13880209.2017.1392987] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 08/21/2017] [Accepted: 09/21/2017] [Indexed: 06/07/2023]
Abstract
CONTEXT The effects of the anticancer drug paclitaxel on learning and memory are rarely studied. OBJECTIVE This study investigated changes in GABAB receptor expression during paclitaxel-induced apoptosis of hippocampal neurons and the role of the p38MAPK/NF-κB pathway in this process. MATERIALS AND METHODS Hippocampal neurons isolated from neonatal Sprague-Dawley rats were divided into six groups: Control (C), SB (10 µL of 10-µmol/L SB203580), SN (53 µg/mL SN50), N (1 µmol/L paclitaxel), SB + N (10 µmol/L SB203580 + 1 µmol/L paclitaxel) and SN + N (53 µg/mL SN50 + 1 µmol/L paclitaxel). Cells in different groups were treated with corresponding agents for 24 h at 37 °C. The apoptosis rate and protein levels of GABAB1 receptors and NF-κB p65 were evaluated. Rat models of neuropathic pain was induced by paclitaxel and were divided into four groups such as N, B + N, SN + N and SN + B + N groups. Rats in the N group received intrathecal injections of normal saline solution. Rats in the B + N group received intrathecal injections of 10 μL baclofen (0.05 μg/μL). Rats in the SN + N and SN + B + N groups received intrathecal injections of SN50 and SN50 plus baclofen, respectively. Spatial learning and memory were evaluated in rat models based on the escape latency and the number of crossings over the platform and protein levels of GABAB1 receptors, NF-κB, IL-1β and TNFα were measured by immunohistochemistry assay and western blot. RESULTS The neuronal apoptosis rate was significantly increased in N (49.16 ± 3.12)%, SB + N (31.18 ± 3.02)% and SN + N (28.47 ± 3.75)% groups, accompanied by increased levels of GABAB1 receptors and NF-κB p65 (p < 0.05). The paclitaxel-treated rats demonstrated significantly increased latency (24.32 ± 2.94)s and decreased the crossings number (3.14 ± 0.63) after 15 d in the Morris water maze (p < 0.05). Immunohistochemistry assay showed that compared with the N group (GABAB1:9.0 ± 1.6, NF-κB p65:29.6 ± 2.4, IL-1β: 30.4 ± 3.4, TNFα: 31.0 ± 3.4), B + N, SN + N and SN + B + N groups evidently increased levels of GABAB1 receptor (B + N:SN + N:SN + B + N = 19.4 ± 2.1:20.8 ± 1.9:28.0 ± 1.9) but significantly decreased levels of NF-κB p65 (B + N:SN + N:SN + B + N = 21.2 ± 1.5:18.6 ± 2.1:12.6 ± 1.5), IL-1β (B + N:SN + N:SN + B + N = 22.0 ± 1.0:19.6 ± 1.8:14.6 ± 1.5) and TNF α (B + N:SN + N:SN + B + N = 23.0 ± 1.6:22.2 ± 0.8:16.6 ± 1.7). Similar findings were found in western blot analysis. DISCUSSIONS AND CONCLUSIONS Paclitaxel may reduce cognitive function in rats through the p38MAPK/NF-κB pathway and GABAB1 receptors.
Collapse
Affiliation(s)
- Zhao Li
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Peng Liu
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Hailin Zhang
- Department of Pharmacology, HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Shuang Zhao
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Zi Jin
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Rui Li
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Yuexian Guo
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| | - Xiuli Wang
- Department of Anesthesiology, The Third Hospital of HeBei Medical University, Shijiazhuang, HeBei Province, China
| |
Collapse
|
9
|
Bonomi A, Ghezzi E, Pascucci L, Aralla M, Ceserani V, Pettinari L, Coccè V, Guercio A, Alessandri G, Parati E, Brini AT, Zeira O, Pessina A. Effect of canine mesenchymal stromal cells loaded with paclitaxel on growth of canine glioma and human glioblastoma cell lines. Vet J 2017; 223:41-47. [PMID: 28671070 DOI: 10.1016/j.tvjl.2017.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 05/03/2017] [Accepted: 05/14/2017] [Indexed: 01/03/2023]
Abstract
This study investigated whether canine mesenchymal stromal cells (cMSCs) are able to take up and release paclitaxel (PTX) in active form, and therefore whether they have potential as a tool for therapeutic delivery of this drug. cMSCs from bone marrow and adipose tissue were isolated, expanded and characterised phenotypically. cMSCs were loaded with PTX (cMSCs-PTX) and their capacity for release of PTX was determined by their effect on proliferation of cancer cells. cMSCs-PTX derived from bone marrow and adipose tissue were able to take up and then release active PTX. cMSCs-PTC inhibited proliferation of the canine glioma cell line J3T, and the human glioblastoma cell lines T98G and U87MG. The potential of canine cMSCs-PTX for treatment of canine gliomas should be investigated further.
Collapse
Affiliation(s)
- A Bonomi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, via Pascal 36, 20133 Milan, Italy
| | - E Ghezzi
- Veterinary Hospital San Michele, via I Maggio 37, 26838 Tavazzano con Villavesco, Lodi, Italy
| | - L Pascucci
- Department of Veterinary Medicine, University of Perugia, via San Costanzo 4, 06126 Perugia, Italy
| | - M Aralla
- Veterinary Hospital San Michele, via I Maggio 37, 26838 Tavazzano con Villavesco, Lodi, Italy
| | - V Ceserani
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, via Celoria 11, 20133 Milan, Italy
| | - L Pettinari
- Veterinary Hospital San Michele, via I Maggio 37, 26838 Tavazzano con Villavesco, Lodi, Italy
| | - V Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, via Pascal 36, 20133 Milan, Italy
| | - A Guercio
- Area Diagnostica Virologica dell'Istituto Zooprofilattico Sperimentale della Sicilia 'A Mirri', Palermo, Italy
| | - G Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, via Celoria 11, 20133 Milan, Italy
| | - E Parati
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, via Celoria 11, 20133 Milan, Italy
| | - A T Brini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, via Pascal 36, 20133 Milan, Italy; IRCCS Galeazzi Orthopaedic Institute, Milan, Italy
| | - O Zeira
- Veterinary Hospital San Michele, via I Maggio 37, 26838 Tavazzano con Villavesco, Lodi, Italy
| | - A Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, via Pascal 36, 20133 Milan, Italy.
| |
Collapse
|
10
|
Schiariti MP, Restelli F, Ferroli P, Benetti A, Berenzi A, Ferri A, Ceserani V, Ciusani E, Cadei M, Finocchiaro G, Pessina A, Parati E, Pallini R, Alessandri G. Fibronectin-adherent peripheral blood derived mononuclear cells as Paclitaxel carriers for glioblastoma treatment: An in vitro study. Cytotherapy 2017; 19:721-734. [PMID: 28434806 DOI: 10.1016/j.jcyt.2017.03.069] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/20/2017] [Accepted: 03/10/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Glioblastoma (GBM) represents the most aggressive malignant brain tumor in adults, with a risible median life expectancy despite gold standard treatment. Novel drug-delivery methods have been explored. Here we evaluated the possibility to use mononuclear cells (MCs) belonging to the monocytic-dendritic lineage as drug-carrier. METHODS MCs were obtained from 10 patients harboring a GBM, and from healthy volunteers, considered as controls. GBM tissue was also obtained from patients. MCs were cultured and the adherent population on fibronectin (FN-MCs), after immunocytochemistry and flow cytometry characterization, was loaded with Paclitaxel (FN-MCs-PTX). Antiproliferative and migration activity of FN-MCs-PTX was evaluated in two-dimensional (2D) and three-dimensional (3D) co-culture assays with red fluorescent U87 Malignant Glioma cells and primary GBM cells. Antiangiogenic properties of FN-MCs-PTX were tested on cultures with endothelial cells. RESULTS Phenotypical characterization showed a high expression of monocytic-dendritic markers in GBM cells and FN-MCs. FN-MCs demonstrated to effectively uptake PTX and to strongly inhibit GBM growth in vitro (P <0.01). Moreover, tumor-induced migration of MCs, although partially affected by the PTX cargo, remained statistically significant when compared with unprimed cells and this was confirmed in a 3D Matrigel model (P <0.01) and in a Trans-well assay (P <0.01). FN-MCs-PTX also disclosed considerable antiangiogenic properties. DISCUSSION Our results suggest that the fibronectin-adherent population of MCs isolated from peripheral blood can be an effective tool to inhibit GBM growth. Given the relative facility to obtain such cells and the short time needed for their culture and drug loading this approach may have potential as an adjuvant therapy for GBM.
Collapse
Affiliation(s)
- Marco Paolo Schiariti
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.
| | - Francesco Restelli
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Paolo Ferroli
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Anna Benetti
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Angiola Berenzi
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Anna Ferri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Valentina Ceserani
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Emilio Ciusani
- Laboratory of Clinical Pathology and Neurogenetic Medicine, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Moris Cadei
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Gaetano Finocchiaro
- Molecular Neuroncology Unit, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Eugenio Parati
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Roberto Pallini
- Institute of Neurosurgery, Catholic University of Sacro Cuore, Roma, Italy
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| |
Collapse
|
11
|
Girdlestone J. Mesenchymal stromal cells with enhanced therapeutic properties. Immunotherapy 2016; 8:1405-1416. [DOI: 10.2217/imt-2016-0098] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
12
|
Emerging role of mesenchymal stem cells during tuberculosis: The fifth element in cell mediated immunity. Tuberculosis (Edinb) 2016; 101S:S45-S52. [DOI: 10.1016/j.tube.2016.09.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Pessina A, Leonetti C, Artuso S, Benetti A, Dessy E, Pascucci L, Passeri D, Orlandi A, Berenzi A, Bonomi A, Coccè V, Ceserani V, Ferri A, Dossena M, Mazzuca P, Ciusani E, Ceccarelli P, Caruso A, Portolani N, Sisto F, Parati E, Alessandri G. Drug-releasing mesenchymal cells strongly suppress B16 lung metastasis in a syngeneic murine model. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:82. [PMID: 26264809 PMCID: PMC4534150 DOI: 10.1186/s13046-015-0200-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are considered an important therapeutic tool in cancer therapy. They possess intrinsic therapeutic potential and can also be in vitro manipulated and engineered to produce therapeutic molecules that can be delivered to the site of diseases, through their capacity to home pathological tissues. We have recently demonstrated that MSCs, upon in vitro priming with anti-cancer drug, become drug-releasing mesenchymal cells (Dr-MCs) able to strongly inhibit cancer cells growth. METHODS Murine mesenchymal stromal cells were loaded with Paclitaxel (Dr-MCsPTX) according to a standardized procedure and their ability to inhibit the growth of a murine B16 melanoma was verified by in vitro assays. The anti-metastatic activity of Dr-MCsPTX was then studied in mice injected i.v. with B16 melanoma cells that produced lung metastatic nodules. Lung nodules were counted under a dissecting stereomicroscope and metastasis investigated by histological analysis. RESULTS We found that three i.v. injections of Dr-MCsPTX on day 5, 10 and 15 after tumor injection almost completely abolished B16 lung metastasis. Dr-MCsPTX arrested into lung by interacting with endothelium and migrate toward cancer nodule through a complex mechanism involving primarily mouse lung stromal cells (mL-StCs) and SDF-1/CXCR4/CXCR7 axis. CONCLUSIONS Our results show for the first time that Dr-MCsPTX are very effective to inhibit lung metastasis formation. Actually, a cure for lung metastasis in humans is mostly unlikely and we do not know whether a therapy combining engineered MSCs and Dr-MCs may work synergistically. However, we think that our approach using Dr-MCs loaded with PTX may represent a new valid and additive therapeutic tool to fight lung metastases and, perhaps, primary lung cancers in human.
Collapse
Affiliation(s)
- Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, Milan, 20133, Italy.
| | - Carlo Leonetti
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Simona Artuso
- Experimental Chemotherapy Laboratory, Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Benetti
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Enrico Dessy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Daniela Passeri
- Department of Biopathology and Image Diagnostics, Anatomic Pathology Institute, University of Rome 'Tor Vergata', Rome, Italy
| | - Augusto Orlandi
- Department of Biopathology and Image Diagnostics, Anatomic Pathology Institute, University of Rome 'Tor Vergata', Rome, Italy
| | - Angiola Berenzi
- Department of Clinical and Experimental Sciences, Institute of Pathological Anatomy, University of Brescia, Brescia, Italy
| | - Arianna Bonomi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, Milan, 20133, Italy
| | - Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, Milan, 20133, Italy
| | - Valentina Ceserani
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Anna Ferri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Marta Dossena
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Pietro Mazzuca
- Department of Microbiology, Brescia University, Brescia, Italy
| | - Emilio Ciusani
- Laboratory of Clinical Pathology and Neurogenetic Medicine, Fondazione IRCCS Neurological Institute Carlo Besta, Milan, Italy
| | - Piero Ceccarelli
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Arnaldo Caruso
- Department of Microbiology, Brescia University, Brescia, Italy
| | - Nazario Portolani
- Department of Medical and Surgical Sciences, University of Brescia, Brescia, Italy
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, Milan, 20133, Italy
| | - Eugenio Parati
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, Milan, Italy
| |
Collapse
|