1
|
Khaseb S, Kohansal Vajari M, Soufi Zomorrod M, Rezai Rad M, Ajami M, Ajami M, Sadeghpour S, Atashi A. Effect of fibrin on the expression of adhesion molecules (ICAM-1, ITGAV, and ITGB3) in unrestricted somatic stem cells. Hematol Transfus Cell Ther 2025; 47:103827. [PMID: 40315755 PMCID: PMC12098137 DOI: 10.1016/j.htct.2025.103827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/18/2024] [Accepted: 11/10/2024] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Hematopoietic stem cell expansion relies on direct cell-cell interactions mediated by adhesion molecules, integrins, and cytokines. Unrestricted somatic stem cells have emerged as novel stromal cells supporting hematopoietic stem cell expansion in co-culture conditions via secretion of hematopoiesis-related cytokines and the expression of adhesion molecules. Previous research showed fibrin increased hematopoiesis-related gene expression in these cells. This study focused on the adhesive characteristics of unrestricted somatic stem cells on 3D fibrin scaffolds. METHODS Unrestricted somatic stem cells were isolated from umbilical cord blood and characterized using flow cytometry and multilineage differentiation assays. Scanning electron microscopy and DAPI staining were employed to analyze cell attachment to fibrin. Viability on fibrin was assessed through MTT assays. Quantitative polymerase chain reaction was conducted to evaluate the expression of intercellular adhesion molecule 1 (ICAM-1), integrin subunit αv (ITGAV), and integrin subunit β3 (ITGB3) in cells cultured on 3D fibrin scaffolds. RESULTS Cells were positive for CD73, CD105, and CD166 but negative for CD45. Alizarin red and Oil red O stains confirmed calcium deposition and lipid vacuoles. MTT assays revealed that fibrin positively impacts viability. ITGAV expression was significantly increased in cells cultured on fibrin compared to those cultured on plastic tissue culture plates (Control Group). Furthermore, ITGB3 expression showed no significant change in both groups, while ICAM-1 expression was downregulated in cells cultured on fibrin. CONCLUSIONS Our study revealed that fibrin has a positive impact on the expression of ITGAV, which plays a crucial role in direct cell-cell interactions affecting hematopoietic stem cell expansion.
Collapse
Affiliation(s)
- Sanaz Khaseb
- Faculty of Medical Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| | - Mahdi Kohansal Vajari
- Faculty of Medical Sciences, Tarbiat Modares University (TMU), Tehran, Iran; School of Allied Medical Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Maryam Rezai Rad
- Research Institute for Dental Sciences, Dental Research Center, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Monireh Ajami
- Faculty of Paramedical Sciences, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mansoureh Ajami
- School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Saba Sadeghpour
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
2
|
King JL, Valdivia A, Hingtgen SD, Benhabbour SR. Injectable Tumoricidal Neural Stem Cell-Laden Hydrogel for Treatment of Glioblastoma Multiforme-An In Vivo Safety, Persistence, and Efficacy Study. Pharmaceutics 2024; 17:3. [PMID: 39861656 PMCID: PMC11768746 DOI: 10.3390/pharmaceutics17010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Glioblastoma multiforme (GBM) is the most common high-grade primary brain cancer in adults. Despite efforts to advance treatment, GBM remains treatment resistant and inevitably progresses after first-line therapy. Induced neural stem cell (iNSC) therapy is a promising, personalized cell therapy approach that has been explored to circumvent challenges associated with the current GBM treatment. METHODS Herein, we developed a chitosan-based (CS) injectable, biodegradable, in situ forming thermo-responsive hydrogel as a cell delivery vehicle for the treatment of GBM. Tumoricidal neural stem cells were encapsulated in the injectable CS hydrogel as stem cell therapy for treatment of post-surgical GBM. In this report, we investigated the safety of the injectable CS hydrogel in an immune-competent mouse model. Furthermore, we evaluated the persistence and efficacy of iNSC-laden CS hydrogels in a post-surgical GBM mouse model. RESULTS The injectable CS hydrogel was well tolerated in mice with no signs of chronic local inflammation. Induced neural stem cells (iNSCs) persisted in the CS hydrogels for over 196 days in comparison to 21 days for iNSCs (cell injection) only. GBM recurrence was significantly slower in mice treated with iNSC-laden CS hydrogels with a 50% increase in overall median survival in comparison to iNSCs (cell injection) only. CONCLUSIONS Collectively, we demonstrated the ability to encapsulate, retain, and deliver iNSCs in an injectable CS hydrogel that is well tolerated with better survival rates than iNSCs alone.
Collapse
Affiliation(s)
- Jasmine L. King
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA;
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.V.); (S.D.H.)
| | - Shawn D. Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.V.); (S.D.H.)
| | - S. Rahima Benhabbour
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.V.); (S.D.H.)
| |
Collapse
|
3
|
Al‐Kharboosh R, Bechtle A, Tzeng SY, Zheng J, Mondal SK, Wilson DR, Perez‐Vega C, Green JJ, Quiñones‐Hinojosa A. Therapeutic potential and impact of nanoengineered patient-derived mesenchymal stem cells in a murine resection and recurrence model of human glioblastoma. Bioeng Transl Med 2024; 9:e10675. [PMID: 39545093 PMCID: PMC11558202 DOI: 10.1002/btm2.10675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 11/17/2024] Open
Abstract
Confounding results of engineered mesenchymal stem cells (MSCs) used as cellular vehicles has plagued technologies whereby success or failure of novel approaches may be dismissed or inaccurately ascribed solely to the biotechnology platform rather than suitability of the human donor. Polymeric materials were screened for non-viral engineering of MSCs from multiple human donors to deliver bone morphogenic protein-4 (BMP4), a protein previously investigated in clinical trials for glioblastoma (GBM) to combat a subpopulation of highly invasive and tumorigenic clones. A "smart technology" that target the migratory and stem-like nature of GBM will require: (1) a cellular vehicle (MSC) which can scavenge and target residual cells left behind after surgical debulking and deliver; (2) anti-glioma cargo (BMP4). Multiple MSC donors are safely engineered, though varied in susceptibility to accept BMP4 due to intrinsic characteristics revealed by their molecular signatures. Efficiency is compared via secretion, downstream signaling, differentiation, and anti-proliferative properties across all donors. In a clinically relevant resection and recurrence model of patient-derived human GBM, we demonstrate that nanoengineered MSCs are not "donor agnostic" and efficacy is influenced by the inherent suitability of the MSC to the cargo. Therefore, donor profiles hold greater influence in determining downstream outcomes than the technical capabilities of the engineering technology.
Collapse
Affiliation(s)
- Rawan Al‐Kharboosh
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
- Department of NeuroscienceMayo Clinic Graduate SchoolJacksonvilleFloridaUSA
- AtPoint tx Co.WashingtonDistrict of ColumbiaUSA
| | - Alex Bechtle
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
| | - Stephany Y. Tzeng
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jiaying Zheng
- Department of NeuroscienceMayo Clinic Graduate SchoolJacksonvilleFloridaUSA
| | | | - David R. Wilson
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Jordan J. Green
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Departments of Neurosurgery, Oncology, Ophthalmology, Materials Science & Engineering, and Chemical & Biomolecular EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | |
Collapse
|
4
|
Mei J, Jiang XY, Tian HX, Rong DC, Song JN, Wang L, Chen YS, Wong RCB, Guo CX, Wang LS, Wang LY, Wang PY, Yin JY. Anoikis in cell fate, physiopathology, and therapeutic interventions. MedComm (Beijing) 2024; 5:e718. [PMID: 39286778 PMCID: PMC11401975 DOI: 10.1002/mco2.718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
The extracellular matrix (ECM) governs a wide spectrum of cellular fate processes, with a particular emphasis on anoikis, an integrin-dependent form of cell death. Currently, anoikis is defined as an intrinsic apoptosis. In contrast to traditional apoptosis and necroptosis, integrin correlates ECM signaling with intracellular signaling cascades, describing the full process of anoikis. However, anoikis is frequently overlooked in physiological and pathological processes as well as traditional in vitro research models. In this review, we summarized the role of anoikis in physiological and pathological processes, spanning embryonic development, organ development, tissue repair, inflammatory responses, cardiovascular diseases, tumor metastasis, and so on. Similarly, in the realm of stem cell research focused on the functional evolution of cells, anoikis offers a potential solution to various challenges, including in vitro cell culture models, stem cell therapy, cell transplantation, and engineering applications, which are largely based on the regulation of cell fate by anoikis. More importantly, the regulatory mechanisms of anoikis based on molecular processes and ECM signaling will provide new strategies for therapeutic interventions (drug therapy and cell-based therapy) in disease. In summary, this review provides a systematic elaboration of anoikis, thus shedding light on its future research.
Collapse
Affiliation(s)
- Jie Mei
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Xue-Yao Jiang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Hui-Xiang Tian
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Ding-Chao Rong
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
| | - Jia-Nan Song
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- School of Life Sciences Westlake University Hangzhou Zhejiang China
| | - Luozixian Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Yuan-Shen Chen
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Raymond C B Wong
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Cheng-Xian Guo
- Center of Clinical Pharmacology the Third Xiangya Hospital Central South University Changsha Hunan China
| | - Lian-Sheng Wang
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Lei-Yun Wang
- Department of Pharmacy Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan Hubei Province China
| | - Peng-Yuan Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| |
Collapse
|
5
|
Cai ZM, Li ZZ, Zhong NN, Cao LM, Xiao Y, Li JQ, Huo FY, Liu B, Xu C, Zhao Y, Rao L, Bu LL. Revolutionizing lymph node metastasis imaging: the role of drug delivery systems and future perspectives. J Nanobiotechnology 2024; 22:135. [PMID: 38553735 PMCID: PMC10979629 DOI: 10.1186/s12951-024-02408-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
The deployment of imaging examinations has evolved into a robust approach for the diagnosis of lymph node metastasis (LNM). The advancement of technology, coupled with the introduction of innovative imaging drugs, has led to the incorporation of an increasingly diverse array of imaging techniques into clinical practice. Nonetheless, conventional methods of administering imaging agents persist in presenting certain drawbacks and side effects. The employment of controlled drug delivery systems (DDSs) as a conduit for transporting imaging agents offers a promising solution to ameliorate these limitations intrinsic to metastatic lymph node (LN) imaging, thereby augmenting diagnostic precision. Within the scope of this review, we elucidate the historical context of LN imaging and encapsulate the frequently employed DDSs in conjunction with a variety of imaging techniques, specifically for metastatic LN imaging. Moreover, we engage in a discourse on the conceptualization and practical application of fusing diagnosis and treatment by employing DDSs. Finally, we venture into prospective applications of DDSs in the realm of LNM imaging and share our perspective on the potential trajectory of DDS development.
Collapse
Affiliation(s)
- Ze-Min Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Jia-Qi Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Fang-Yi Huo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
- Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, Hubei, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, Brisbane, QLD, 4066, Australia
| | - Yi Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China.
- Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, Hubei, China.
| |
Collapse
|
6
|
Charvátová S, Motais B, Czapla J, Cichoń T, Smolarczyk R, Walek Z, Giebel S, Hájek R, Bagó JR. Novel Local "Off-the-Shelf" Immunotherapy for the Treatment of Myeloma Bone Disease. Cells 2023; 12:cells12030448. [PMID: 36766789 PMCID: PMC9914109 DOI: 10.3390/cells12030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/13/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Myeloma bone disease (MBD) is one of the major complications in multiple myeloma (MM)-the second most frequent hematologic malignancy. It is characterized by the formation of bone lesions due to the local action of proliferating MM cells, and to date, no effective therapy has been developed. In this study, we propose a novel approach for the local treatment of MBD with a combination of natural killer cells (NKs) and mesenchymal stem cells (MSCs) within a fibrin scaffold, altogether known as FINM. The unique biological properties of the NKs and MSCs, joined to the injectable biocompatible fibrin, permitted to obtain an efficient "off-the-shelf" ready-to-use composite for the local treatment of MBD. Our in vitro analyses demonstrate that NKs within FINM exert a robust anti-tumor activity against MM cell lines and primary cells, with the capacity to suppress osteoclast activity (~60%) within in vitro 3D model of MBD. Furthermore, NKs' post-thawing cytotoxic activity is significantly enhanced (~75%) in the presence of MSCs, which circumvents the decrease of NKs cytotoxicity after thawing, a well-known issue in the cryopreservation of NKs. To reduce the tumor escape, we combined FINM with other therapeutic agents (bortezomib (BZ), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)), observing a clear therapeutic synergistic effect in vitro. Finally, the therapeutic efficacy of FINM in combination with BZ and TRAIL was assessed in a mouse model of MM, achieving 16-fold smaller tumors compared to the control group without treatment. These results suggest the potential of FINM to serve as an allogeneic "off-the-shelf" approach to improve the outcomes of patients suffering from MBD.
Collapse
Affiliation(s)
- Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Benjamin Motais
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, 70100 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Justyna Czapla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Tomasz Cichoń
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Ryszard Smolarczyk
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Zuzana Walek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Sebastian Giebel
- Department of Bone Marrow Transplantation and Onco-Hematology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44102 Gliwice, Poland
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| | - Juli R. Bagó
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 70800 Ostrava, Czech Republic
- Correspondence: ; Tel.: +42-(05)-97372092
| |
Collapse
|
7
|
King JL, Maturavongsadit P, Hingtgen SD, Benhabbour SR. Injectable pH Thermo-Responsive Hydrogel Scaffold for Tumoricidal Neural Stem Cell Therapy for Glioblastoma Multiforme. Pharmaceutics 2022; 14:pharmaceutics14102243. [PMID: 36297678 PMCID: PMC9609352 DOI: 10.3390/pharmaceutics14102243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common malignant brain tumor in adults and despite recent advances in treatment modalities, GBM remains incurable. Injectable hydrogel scaffolds are a versatile delivery system that can improve delivery of drug and cell therapeutics for GBM. In this report, we investigated an injectable nanocellulose/chitosan-based hydrogel scaffold for neural stem cell encapsulation and delivery. Hydrogels were prepared using thermogelling beta-glycerophosphate (BGP) and hydroxyethyl cellulose (HEC), chitosan (CS), and cellulose nanocrystals (CNCs). We evaluated the impact of neural stem cells on hydrogel gelation kinetics, microstructures, and degradation. Furthermore, we investigated the biomaterial effects on cell viability and functionality. We demonstrated that the incorporation of cells at densities of 1, 5 and 10 million does not significantly impact rheological and physical properties CS scaffolds. However, addition of CNCs significantly prolonged hydrogel degradation when cells were seeded at 5 and 10 million per 1 mL hydrogel. In vitro cell studies demonstrated high cell viability, release of TRAIL at therapeutic concentrations, and effective tumor cell killing within 72 h. The ability of these hydrogel scaffolds to support stem cell encapsulation and viability and maintain stem cell functionality makes them an attractive cell delivery system for local treatment of post-surgical cancers.
Collapse
Affiliation(s)
- Jasmine L. King
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Panita Maturavongsadit
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Shawn D. Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - S. Rahima Benhabbour
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
- Correspondence:
| |
Collapse
|
8
|
Bomba HN, Carey‐Ewend A, Sheets KT, Valdivia A, Goetz M, Findlay IA, Mercer‐Smith A, Kass LE, Khagi S, Hingtgen SD. Use of
FLOSEAL
® as a scaffold and its impact on induced neural stem cell phenotype, persistence, and efficacy. Bioeng Transl Med 2022; 7:e10283. [PMID: 35600639 PMCID: PMC9115686 DOI: 10.1002/btm2.10283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 01/15/2023] Open
Abstract
Induced neural stem cells (iNSCs) have emerged as a promising therapeutic platform for glioblastoma (GBM). iNSCs have the innate ability to home to tumor foci, making them ideal carriers for antitumor payloads. However, the in vivo persistence of iNSCs limits their therapeutic potential. We hypothesized that by encapsulating iNSCs in the FDA‐approved, hemostatic matrix FLOSEAL®, we could increase their persistence and, as a result, therapeutic durability. Encapsulated iNSCs persisted for 95 days, whereas iNSCs injected into the brain parenchyma persisted only 2 weeks in mice. Two orthotopic GBM tumor models were used to test the efficacy of encapsulated iNSCs. In the GBM8 tumor model, mice that received therapeutic iNSCs encapsulated in FLOSEAL® survived 30 to 60 days longer than mice that received nonencapsulated cells. However, the U87 tumor model showed no significant differences in survival between these two groups, likely due to the more solid and dense nature of the tumor. Interestingly, the interaction of iNSCs with FLOSEAL® appears to downregulate some markers of proliferation, anti‐apoptosis, migration, and therapy which could also play a role in treatment efficacy and durability. Our results demonstrate that while FLOSEAL® significantly improves iNSC persistence, this alone is insufficient to enhance therapeutic durability.
Collapse
Affiliation(s)
- Hunter N. Bomba
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Abigail Carey‐Ewend
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Kevin T. Sheets
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Alain Valdivia
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Morgan Goetz
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Ingrid A. Findlay
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Alison Mercer‐Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Lauren E. Kass
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Simon Khagi
- Department of Neurosurgery The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Shawn D. Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Lineberger Comprehensive Cancer Center The University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| |
Collapse
|
9
|
King JL, Benhabbour SR. Glioblastoma Multiforme-A Look at the Past and a Glance at the Future. Pharmaceutics 2021; 13:pharmaceutics13071053. [PMID: 34371744 PMCID: PMC8309001 DOI: 10.3390/pharmaceutics13071053] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 01/14/2023] Open
Abstract
Gliomas are the most common type of brain tumor that occur in adults and children. Glioblastoma multiforme (GBM) is the most common, aggressive form of brain cancer in adults and is universally fatal. The current standard-of-care options for GBM include surgical resection, radiotherapy, and concomitant and/or adjuvant chemotherapy. One of the major challenges that impedes success of chemotherapy is the presence of the blood–brain barrier (BBB). Because of the tightly regulated BBB, immune surveillance in the central nervous system (CNS) is poor, contributing to unregulated glioma cell growth. This review gives a comprehensive overview of the latest advances in treatment of GBM with emphasis on the significant advances in immunotherapy and novel therapeutic delivery strategies to enhance treatment for GBM.
Collapse
Affiliation(s)
- Jasmine L. King
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Soumya Rahima Benhabbour
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +1-919-843-6142
| |
Collapse
|
10
|
King JL, Benhabbour SR. Glioblastoma Multiforme-A Look at the Past and a Glance at the Future. Pharmaceutics 2021; 13:1053. [PMID: 34371744 PMCID: PMC8309001 DOI: 10.3390/pharmaceutics13071053;] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Gliomas are the most common type of brain tumor that occur in adults and children. Glioblastoma multiforme (GBM) is the most common, aggressive form of brain cancer in adults and is universally fatal. The current standard-of-care options for GBM include surgical resection, radiotherapy, and concomitant and/or adjuvant chemotherapy. One of the major challenges that impedes success of chemotherapy is the presence of the blood-brain barrier (BBB). Because of the tightly regulated BBB, immune surveillance in the central nervous system (CNS) is poor, contributing to unregulated glioma cell growth. This review gives a comprehensive overview of the latest advances in treatment of GBM with emphasis on the significant advances in immunotherapy and novel therapeutic delivery strategies to enhance treatment for GBM.
Collapse
Affiliation(s)
- Jasmine L. King
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Soumya Rahima Benhabbour
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +1-919-843-6142
| |
Collapse
|
11
|
Engineered microtissues for the bystander therapy against cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111854. [PMID: 33579487 DOI: 10.1016/j.msec.2020.111854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022]
Abstract
Thymidine kinase expressing human adipose mesenchymal stem cells (TK-hAMSCs) in combination with ganciclovir (GCV) are an effective platform for antitumor bystander therapy in mice models. However, this strategy requires multiple TK-hAMSCs administrations and a substantial number of cells. Therefore, for clinical translation, it is necessary to find a biocompatible scaffold providing TK-hAMSCs retention in the implantation site against their rapid wash-out. We have developed a microtissue (MT) composed by TKhAMSCs and a scaffold made of polylactic acid microparticles and cell-derived extracellular matrix deposited by hAMSCs. The efficacy of these MTs as vehicles for TK-hAMSCs/GCV bystander therapy was evaluated in a rodent model of human prostate cancer. Subcutaneously implanted MTs were integrated in the surrounding tissue, allowing neovascularization and maintenance of TK-hAMSCs viability. Furthermore, MTs implanted beside tumors allowed TK-hAMSCs migration towards tumor cells and, after GCV administration, inhibited tumor growth. These results indicate that TK-hAMSCs-MTs are promising cell reservoirs for clinical use of therapeutic MSCs in bystander therapies.
Collapse
|
12
|
Tang M, Rich JN, Chen S. Biomaterials and 3D Bioprinting Strategies to Model Glioblastoma and the Blood-Brain Barrier. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004776. [PMID: 33326131 PMCID: PMC7854518 DOI: 10.1002/adma.202004776] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/06/2020] [Indexed: 05/13/2023]
Abstract
Glioblastoma (GBM) is the most prevalent and lethal adult primary central nervous system cancer. An immunosuppresive and highly heterogeneous tumor microenvironment, restricted delivery of chemotherapy or immunotherapy through the blood-brain barrier (BBB), together with the brain's unique biochemical and anatomical features result in its universal recurrence and poor prognosis. As conventional models fail to predict therapeutic efficacy in GBM, in vitro 3D models of GBM and BBB leveraging patient- or healthy-individual-derived cells and biomaterials through 3D bioprinting technologies potentially mimic essential physiological and pathological features of GBM and BBB. 3D-bioprinted constructs enable investigation of cellular and cell-extracellular matrix interactions in a species-matched, high-throughput, and reproducible manner, serving as screening or drug delivery platforms. Here, an overview of current 3D-bioprinted GBM and BBB models is provided, elaborating on the microenvironmental compositions of GBM and BBB, relevant biomaterials to mimic the native tissues, and bioprinting strategies to implement the model fabrication. Collectively, 3D-bioprinted GBM and BBB models are promising systems and biomimetic alternatives to traditional models for more reliable mechanistic studies and preclinical drug screenings that may eventually accelerate the drug development process for GBM.
Collapse
Affiliation(s)
- Min Tang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jeremy N. Rich
- Division of Regenerative Medicine, Department of Medicine, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, Materials Science and Engineering Program, Chemical Engineering Program, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
13
|
Zhang Y, Jiang C. Postoperative cancer treatments: In-situ delivery system designed on demand. J Control Release 2021; 330:554-564. [PMID: 33359583 DOI: 10.1016/j.jconrel.2020.12.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
The keys to the prevention of tumor recurrence after operation are the elimination of residual tumor cells and the reversal of microenvironments that induce recurrence. In the formulation of a treatment scheme, building an appropriate drug delivery system is essential. An in-situ drug delivery system (ISDDS) is regarded as an effective treatment route for postoperative use that increases drug delivery efficiency and mitigates side-effects. ISDDS technology has been considerably improved through a clearer understanding of the mechanisms of postoperative recurrence and the development of drug delivery materials. This paper describes the initiation and characteristics of postoperative recurrence mechanisms. Based on this information, design principles for ISDDS are proposed, and a variety of practical drug delivery systems that fulfil specific therapeutic needs are presented. Challenges and future opportunities related to the application of in-situ drug carriers for inhibiting cancer recurrence are also discussed.
Collapse
Affiliation(s)
- Yiwen Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
14
|
Moore KM, Murthy AB, Graham-Gurysh EG, Hingtgen SD, Bachelder EM, Ainslie KM. Polymeric Biomaterial Scaffolds for Tumoricidal Stem Cell Glioblastoma Therapy. ACS Biomater Sci Eng 2020; 6:3762-3777. [PMID: 33463324 PMCID: PMC10373914 DOI: 10.1021/acsbiomaterials.0c00477] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) is the most common primary brain tumor and has a poor prognosis; as such, there is an urgent need to develop innovative new therapies. Tumoricidal stem cells are an emerging therapy that has the potential to combat limitations of traditional local and systemic chemotherapeutic strategies for GBM by providing a source for high, sustained concentrations of tumoricidal agents locally to the tumor. One major roadblock for tumoricidal stem cell therapy is that the persistence of tumoricidal stem cells injected as a cell suspension into the GBM surgical resection cavity is limited. Polymeric biomaterial scaffolds have been utilized to enhance the delivery of tumoricidal stem cells in the surgical resection cavity and extend their persistence in the brain, ultimately increasing their therapeutic efficacy against GBM. In this review, we examine three main scaffold categories explored for tumoricidal stem cell therapy: microcapsules, hydrogels, and electrospun scaffolds. Furthermore, considering the significant impact of surgery on the brain and recurrent GBM, we survey a brief history of orthotopic models of GBM surgical resection.
Collapse
Affiliation(s)
- Kathryn M Moore
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27599, United States
| | - Ananya B Murthy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Elizabeth G Graham-Gurysh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Kristy M Ainslie
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina 27599, United States.,Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States.,Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
15
|
Motais B, Charvátová S, Hrdinka M, Šimíček M, Jelínek T, Ševčíková T, Kořístek Z, Hájek R, Bagó JR. A Bird's-Eye View of Cell Sources for Cell-Based Therapies in Blood Cancers. Cancers (Basel) 2020; 12:E1333. [PMID: 32456165 PMCID: PMC7281611 DOI: 10.3390/cancers12051333] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/25/2022] Open
Abstract
: Hematological malignancies comprise over a hundred different types of cancers and account for around 6.5% of all cancers. Despite the significant improvements in diagnosis and treatment, many of those cancers remain incurable. In recent years, cancer cell-based therapy has become a promising approach to treat those incurable hematological malignancies with striking results in different clinical trials. The most investigated, and the one that has advanced the most, is the cell-based therapy with T lymphocytes modified with chimeric antigen receptors. Those promising initial results prepared the ground to explore other cell-based therapies to treat patients with blood cancer. In this review, we want to provide an overview of the different types of cell-based therapies in blood cancer, describing them according to the cell source.
Collapse
Affiliation(s)
- Benjamin Motais
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Sandra Charvátová
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
| | - Matouš Hrdinka
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Michal Šimíček
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tomáš Jelínek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Tereza Ševčíková
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Faculty of Science, University of Ostrava, 701 03 Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Zdeněk Kořístek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Roman Hájek
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| | - Juli R. Bagó
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; (B.M.); (S.C.); (M.H.); (M.Š.); (T.J.); (T.Š.); (Z.K.); (R.H.)
- Department of Haematooncology, University Hospital Ostrava, 708 52 Ostrava, Czech Republic
| |
Collapse
|
16
|
Generation and Profiling of Tumor-Homing Induced Neural Stem Cells from the Skin of Cancer Patients. Mol Ther 2020; 28:1614-1627. [PMID: 32402245 DOI: 10.1016/j.ymthe.2020.04.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 03/13/2020] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
The conversion of human fibroblasts into personalized induced neural stem cells (iNSCs) that actively seek out tumors and deliver cytotoxic agents is a highly promising approach for treating various types of cancer. However, the ability to generate iNSCs from the skin of cancer patients has not been explored. Here, we take an important step toward clinical application by generating iNSCs from skin biopsies of human patients undergoing treatment for the aggressive brain cancer, glioblastoma (GBM). We then utilized a panel of functional and genomic studies to investigate the efficacy and tumor-homing capacity of these patient-derived cells, as well as genomic analysis, to characterize the impact of interpatient variability on this personalized cell therapy. From the skin-tissue biopsies, we established fibroblasts and transdifferentiated the cells into iNSCs. Genomic and functional testing revealed marked variability in growth rates, therapeutic agent production, and gene expression during fibroblast-to-iNSC conversion among patient lines. In vivo testing showed patient-derived iNSCs home to tumors, yet rates and expression of homing-related pathways varied among patients. With the use of surgical-resection mouse models of invasive human cluster of differentiation 133+ (CD133+) GBM cells and serial kinetic imaging, we found that "high-performing" patient-derived iNSC lines reduced the volume of GBM cells 60-fold and extended survival from 28 to 45 days. Treatment with "low-performing" patient lines had minimal effect on tumor growth, but the anti-tumor effect could be rescued by increasing the intracavity dose. Together, these data show, for the first time, that tumor-homing iNSCs can be generated from the skin of cancer patients and efficaciously suppress tumor growth. We also begin to define genetic markers that could be used to identify cells that will contain the most effective attributes for tumor homing and kill in human patients, including high gene expression of the semaphorin-3B (SEMA3B), which is known to be involved in neuronal cell migration. These studies should serve as an important guide toward clinical GBM therapy, where the personalized nature of optimized iNSC therapy has the potential to avoid transplant rejection and maximize treatment durability.
Collapse
|
17
|
Moore KM, Graham-Gurysh EG, Bomba HN, Murthy AB, Bachelder EM, Hingtgen SD, Ainslie KM. Impact of composite scaffold degradation rate on neural stem cell persistence in the glioblastoma surgical resection cavity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 111:110846. [PMID: 32279815 DOI: 10.1016/j.msec.2020.110846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/12/2020] [Indexed: 10/24/2022]
Abstract
Tumoricidal neural stem cells (NSCs) are an emerging therapy to combat glioblastoma (GBM). This therapy employs genetically engineered NSCs that secrete tumoricidal agents to seek out and kill tumor foci remaining after GBM surgical resection. Biomaterial scaffolds have previously been utilized to deliver NSCs to the resection cavity. Here, we investigated the impact of scaffold degradation rate on NSC persistence in the brain resection cavity. Composite acetalated dextran (Ace-DEX) gelatin electrospun scaffolds were fabricated with two distinct degradation profiles created by changing the ratio of cyclic to acyclic acetal coverage of Ace-DEX. In vitro, fast degrading scaffolds were fully degraded by one week, whereas slow degrading scaffolds had a half-life of >56 days. The scaffolds also retained distinct degradation profiles in vivo. Two different NSC lines readily adhered to and remained viable on Ace-DEX gelatin scaffolds, in vitro. Therapeutic NSCs secreting tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) had the same TRAIL output as tissue culture treated polystyrene (TCPS) when seeded on both scaffolds. Furthermore, secreted TRAIL was found to be highly potent against the human derived GBM cell line, GBM8, in vitro. Firefly luciferase expressing NSCs were seeded on scaffolds, implanted in a surgical resection cavity and their persistence in the brain was monitored by bioluminescent imaging (BLI). NSC loaded scaffolds were compared to a direct injection (DI) of NSCs in suspension, which is the current clinical approach to NSC therapy for GBM. Fast and slow degrading scaffolds enhanced NSC implantation efficiency 2.87 and 3.08-fold over DI, respectively. Interestingly, scaffold degradation profile did not significantly impact NSC persistence. However, persistence and long-term survival of NSCs was significantly greater for both scaffolds compared to DI, with scaffold implanted NSCs still detected by BLI at day 120 in most mice. Overall, these results highlight the benefit of utilizing a scaffold for application of tumoricidal NSC therapy for GBM.
Collapse
Affiliation(s)
- Kathryn M Moore
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Elizabeth G Graham-Gurysh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Hunter N Bomba
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Ananya B Murthy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Department of Neurosurgery, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Kristy M Ainslie
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
18
|
Developing Implantable Scaffolds to Enhance Neural Stem Cell Therapy for Post-Operative Glioblastoma. Mol Ther 2020; 28:1056-1067. [PMID: 32109370 DOI: 10.1016/j.ymthe.2020.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/17/2020] [Accepted: 02/08/2020] [Indexed: 12/14/2022] Open
Abstract
Pre-clinical and clinical studies have shown that engineered tumoricidal neural stem cells (tNSCs) are a promising treatment strategy for the aggressive brain cancer glioblastoma (GBM). Yet, stabilizing human tNSCs within the surgical cavity following GBM resection is a significant challenge. As a critical step toward advancing engineered human NSC therapy for GBM, we used a preclinical variant of the clinically utilized NSC line HB1.F3.CD and mouse models of human GBM resection/recurrence to identify a polymeric scaffold capable of maximizing the transplant, persistence, and tumor kill of NSC therapy for post-surgical GBM. Using kinetic bioluminescence imaging, we found that tNSCs delivered into the mouse surgical cavity wall by direct injection persisted only 3 days. We found that delivery of tNSCs into the cavity on nanofibrous electrospun poly-l-lactic acid scaffolds extended tNSC persistence to 8 days. Modifications to fiber surface coating, diameter, and morphology of the scaffold failed to significantly extend tNSC persistence in the cavity. In contrast, tNSCs delivered into the post-operative cavity on gelatin matrices (GEMs) persisted 8-fold longer as compared to direct injection. GEMs remained permissive to tumor-tropic homing, as tNSCs migrated off the scaffolds and into invasive tumor foci both in vitro and in vivo. To mirror envisioned human brain tumor trials, we engineered tNSCs to express the prodrug/enzyme thymidine kinase (tNSCstk) and transplanted the therapeutic cells in the post-operative cavity of mice bearing resected orthotopic patient-derived GBM xenografts. Following administration of the prodrug ganciclovir, residual tumor volumes in mice receiving GEM/tNSCs were reduced by 10-fold at day 35, and median survival was extended from 31 to 46 days. Taken together, these data begin to define design parameters for effective scaffold/tNSC composites and suggest a new approach to maximizing the efficacy of tNSC therapy in human patient trials.
Collapse
|
19
|
Aquib M, Juthi AZ, Farooq MA, Ali MG, Janabi AHW, Bavi S, Banerjee P, Bhosale R, Bavi R, Wang B. Advances in local and systemic drug delivery systems for post-surgical cancer treatment. J Mater Chem B 2020; 8:8507-8518. [DOI: 10.1039/d0tb00987c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Graphical representation of local and systemic drug delivery systems.
Collapse
Affiliation(s)
- Md Aquib
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- People's Republic of China
| | - Ajkia Zaman Juthi
- Department of Biochemistry and Molecular Biology
- School of life Science
- University of Science and Technology of China
- Hefei City
- People's Republic of China
| | - Muhammad Asim Farooq
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- People's Republic of China
| | - Manasik Gumah Ali
- Antibody Engineering Laboratory
- School of Life Science & Technology
- China Pharmaceutical University
- Nanjing
- People's Republic of China
| | | | - Sneha Bavi
- Axiom Market Research and ConsultingTM
- Pune 411007
- India
| | - Parikshit Banerjee
- School of Pharmacy, Faculty of Medicine
- The Chinese University of Hong Kong
- New Territories
- People's Republic of China
| | - Raghunath Bhosale
- School of Chemical Sciences
- Punyashlok Ahilyadevi Holkar Solapur University
- Solapur
- India
| | - Rohit Bavi
- School of Chemical Sciences
- Punyashlok Ahilyadevi Holkar Solapur University
- Solapur
- India
- State Key Laboratory of Natural Medicines
| | - Bo Wang
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing
- People's Republic of China
| |
Collapse
|
20
|
Hashemzadeh H, Allahverdi A, Ghorbani M, Soleymani H, Kocsis Á, Fischer MB, Ertl P, Naderi-Manesh H. Gold Nanowires/Fibrin Nanostructure as Microfluidics Platforms for Enhancing Stem Cell Differentiation: Bio-AFM Study. MICROMACHINES 2019; 11:mi11010050. [PMID: 31906040 PMCID: PMC7019962 DOI: 10.3390/mi11010050] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 02/07/2023]
Abstract
Organ-on-a-chip technology has gained great interest in recent years given its ability to control the spatio-temporal microenvironments of cells and tissues precisely. While physical parameters of the respective niche such as microchannel network sizes, geometric features, flow rates, and shear forces, as well as oxygen tension and concentration gradients, have been optimized for stem cell cultures, little has been done to improve cell-matrix interactions in microphysiological systems. Specifically, detailed research on the effect of matrix elasticity and extracellular matrix (ECM) nanotopography on stem cell differentiation are still in its infancy, an aspect that is known to alter a stem cell’s fate. Although a wide range of hydrogels such as gelatin, collagen, fibrin, and others are available for stem cell chip cultivations, only a limited number of elasticities are generally employed. Matrix elasticity and the corresponding nanotopography are key factors that guide stem cell differentiation. Given this, we investigated the addition of gold nanowires into hydrogels to create a tunable biointerface that could be readily integrated into any organ-on-a-chip and cell chip system. In the presented work, we investigated the matrix elasticity (Young’s modulus, stiffness, adhesive force, and roughness) and nanotopography of gold nanowire loaded onto fibrin hydrogels using the bio-AFM (atomic force microscopy) method. Additionally, we investigated the capacity of human amniotic mesenchymal stem cells (hAMSCs) to differentiate into osteo- and chondrogenic lineages. Our results demonstrated that nanogold structured-hydrogels promoted differentiation of hAMSCs as shown by a significant increase in Collagen I and II production. Additionally, there was enhanced calcium mineralization activity and proteoglycans formation after a cultivation period of two weeks within microfluidic devices.
Collapse
Affiliation(s)
- Hadi Hashemzadeh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran;
| | - Abdollah Allahverdi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran; (A.A.); (M.G.); (H.S.)
| | - Mohammad Ghorbani
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran; (A.A.); (M.G.); (H.S.)
| | - Hossein Soleymani
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran; (A.A.); (M.G.); (H.S.)
| | - Ágnes Kocsis
- Department of Health Science and Biomedicine, Danube University Krems, 3500 Vienna, Austria; (Á.K.); (M.B.F.)
| | - Michael Bernhard Fischer
- Department of Health Science and Biomedicine, Danube University Krems, 3500 Vienna, Austria; (Á.K.); (M.B.F.)
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9, 1060 Vienna, Austria
- Correspondence: (P.E.); (H.N.-M.); Tel.: +43(1)-58801-163605 (H.N.M.)
| | - Hossein Naderi-Manesh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran;
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran; (A.A.); (M.G.); (H.S.)
- Correspondence: (P.E.); (H.N.-M.); Tel.: +43(1)-58801-163605 (H.N.M.)
| |
Collapse
|
21
|
Bu LL, Yan J, Wang Z, Ruan H, Chen Q, Gunadhi V, Bell RB, Gu Z. Advances in drug delivery for post-surgical cancer treatment. Biomaterials 2019; 219:119182. [DOI: 10.1016/j.biomaterials.2019.04.027] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/23/2019] [Accepted: 04/23/2019] [Indexed: 02/08/2023]
|
22
|
Dehqan Niri A, Karimi Zarchi AA, Ghadiri Harati P, Salimi A, Mujokoro B. Tissue engineering scaffolds in the treatment of brain disorders in geriatric patients. Artif Organs 2019; 43:947-960. [DOI: 10.1111/aor.13485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Alireza Dehqan Niri
- Nanobiotechnology Research Center Baqiyatallah University of Medical Sciences Tehran Iran
| | | | - Parisa Ghadiri Harati
- Department of Physiotherapy, School of Rehabilitation Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Ali Salimi
- Nanobiotechnology Research Center Baqiyatallah University of Medical Sciences Tehran Iran
| | - Basil Mujokoro
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
23
|
Sheets KT, Bagó JR, Paulk IL, Hingtgen SD. Image-Guided Resection of Glioblastoma and Intracranial Implantation of Therapeutic Stem Cell-seeded Scaffolds. J Vis Exp 2018. [PMID: 30059037 DOI: 10.3791/57452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma (GBM), the most common and aggressive primary brain cancer, carries a life expectancy of 12-15 months. The short life expectancy is due in part to the inability of the current treatment, consisting of surgical resection followed by radiation and chemotherapy, to eliminate invasive tumor foci. Treatment of these foci may be improved with tumoricidal human mesenchymal stem cells (MSCs). MSCs exhibit potent tumor tropism and can be engineered to express therapeutic proteins that kill tumor cells. Advancements in preclinical models indicate that surgical resection induces premature MSC loss and reduces therapeutic efficacy. Efficacy of MSC treatment can be improved by seeding MSCs on a biodegradable poly(lactic acid) (PLA) scaffold. MSC delivery into the surgical resection cavity on a PLA scaffold restores cell retention, persistence, and tumor killing. To study the effects of MSC-seeded PLA implantation on GBM, an accurate preclinical model is needed. Here we provide a preclinical surgical protocol for image-guided tumor resection of GBM in immune-deficient mice followed by MSC-seeded scaffold implantation. MSCs are engineered with lentiviral constructs to constitutively express and secrete therapeutic TNFα-related apoptosis-inducing ligand (TRAIL) as well as green fluorescent protein (GFP) to allow fluorescent tracking. Similarly, the U87 tumor cells are engineered to express mCherry and firefly luciferase, providing dual fluorescent/luminescent tracking. While currently used for investigating stem cell mediated delivery of therapeutics, this protocol could be modified to investigate the impact of surgical resection on other GBM interventions.
Collapse
Affiliation(s)
- Kevin T Sheets
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Juli R Bagó
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Ivory L Paulk
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill;
| |
Collapse
|
24
|
Okolie O, Irvin DM, Bago JR, Sheets K, Satterlee A, Carey-Ewend AG, Lettry V, Dumitru R, Elton S, Ewend MG, Miller CR, Hingtgen SD. Intra-cavity stem cell therapy inhibits tumor progression in a novel murine model of medulloblastoma surgical resection. PLoS One 2018; 13:e0198596. [PMID: 29990322 PMCID: PMC6038981 DOI: 10.1371/journal.pone.0198596] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 05/22/2018] [Indexed: 12/02/2022] Open
Abstract
Background Cytotoxic neural stem cells (NSCs) have emerged as a promising treatment for Medulloblastoma (MB), the most common malignant primary pediatric brain tumor. The lack of accurate pre-clinical models incorporating surgical resection and tumor recurrence limits advancement in post-surgical MB treatments. Using cell lines from two of the 5 distinct MB molecular sub-groups, in this study, we developed an image-guided mouse model of MB surgical resection and investigate intra-cavity NSC therapy for post-operative MB. Methods Using D283 and Daoy human MB cells engineered to express multi-modality optical reporters, we created the first image-guided resection model of orthotopic MB. Brain-derived NSCs and novel induced NSCs (iNSCs) generated from pediatric skin were engineered to express the pro-drug/enzyme therapy thymidine kinase/ganciclovir, seeded into the post-operative cavity, and used to investigate intra-cavity therapy for post-surgical MB. Results We found that surgery reduced MB volumes by 92%, and the rate of post-operative MB regrowth increased 3-fold compared to pre-resection growth. Real-time imaging showed NSCs rapidly homed to MB, migrating 1.6-fold faster and 2-fold farther in the presence of tumors, and co-localized with MB present in the contra-lateral hemisphere. Seeding of cytotoxic NSCs into the post-operative surgical cavity decreased MB volumes 15-fold and extended median survival 133%. As an initial step towards novel autologous therapy in human MB patients, we found skin-derived iNSCs homed to MB cells, while intra-cavity iNSC therapy suppressed post-surgical tumor growth and prolonged survival of MB-bearing mice by 123%. Conclusions We report a novel image-guided model of MB resection/recurrence and provide new evidence of cytotoxic NSCs/iNSCs delivered into the surgical cavity effectively target residual MB foci.
Collapse
Affiliation(s)
- Onyinyechukwu Okolie
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David M. Irvin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Juli R. Bago
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Kevin Sheets
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Andrew Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Abigail G. Carey-Ewend
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Vivien Lettry
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Raluca Dumitru
- UNC Human Pluripotent Stem Cell Core, Genetics Department, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Scott Elton
- Department of Neurosurgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Matthew G. Ewend
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Neurosurgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - C. Ryan Miller
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Neurology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Shawn D. Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Neurosurgery, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- UNC Neuroscience Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
25
|
Cornelison RC, Munson JM. Perspective on Translating Biomaterials Into Glioma Therapy: Lessons From in vitro Models. FRONTIERS IN MATERIALS 2018; 5:27. [PMID: 30911536 PMCID: PMC6430582 DOI: 10.3389/fmats.2018.00027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Glioblastoma (GBM) is the most common and malignant form of brain cancer. Even with aggressive standard of care, GBM almost always recurs because its diffuse, infiltrative nature makes these tumors difficult to treat. The use of biomaterials is one strategy that has been, and is being, employed to study and overcome recurrence. Biomaterials have been used in GBM in two ways: in vitro as mediums in which to model the tumor microenvironment, and in vivo to sustain release of cytotoxic therapeutics. In vitro systems are a useful platform for studying the effects of drugs and tissue-level effectors on tumor cells in a physiologically relevant context. These systems have aided examination of how glioma cells respond to a variety of natural, synthetic, and semi-synthetic biomaterials with varying substrate properties, biochemical factor presentations, and non-malignant parenchymal cell compositions in both 2D and 3D environments. The current in vivo paradigm is completely different, however. Polymeric implants are simply used to line the post-surgical resection cavities and deliver secondary therapies, offering moderate impacts on survival. Instead, perhaps we can use the data generated from in vitro systems to design novel biomaterial-based treatments for GBM akin to a tissue engineering approach. Here we offer our perspective on the topic, summarizing how biomaterials have been used to identify facets of glioma biology in vitro and discussing the elements that show promise for translating these systems in vivo as new therapies for GBM.
Collapse
Affiliation(s)
- R. Chase Cornelison
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jennifer M. Munson
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
26
|
Norouzi M. Recent advances in brain tumor therapy: application of electrospun nanofibers. Drug Discov Today 2018; 23:912-919. [PMID: 29499377 DOI: 10.1016/j.drudis.2018.02.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/11/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
Despite much effort to treat glioblastoma multiforme (GBM), the median survival of patients has not significantly improved. The high rate of tumor recurrence after tumor resection and the blood-brain barrier (BBB) decrease the treatment efficacy. Local drug delivery at the surgical resection site via implantable electrospun nanofibers not only circumvents the BBB, but can also reduce the rate of tumor recurrence. Nanofibers can provide a sustained release and a high concentration of chemotherapeutics at the tumor vicinity, while decreasing their systemic exposure and toxicity. In another scenario, aligned nanofibers can mimic the topographical features of the brain extracellular matrix (ECM), which can be utilized for in vitro studies on GBM cell migration. This strategy is beneficial to investigate the interactions of tumor cells with the microenvironment which has a dominant role in regulating tumor formation, progression, and metastasis.
Collapse
Affiliation(s)
- Mohammad Norouzi
- Graduate Program of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada; Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada.
| |
Collapse
|
27
|
Bagó JR, Okolie O, Dumitru R, Ewend MG, Parker JS, Werff RV, Underhill TM, Schmid RS, Miller CR, Hingtgen SD. Tumor-homing cytotoxic human induced neural stem cells for cancer therapy. Sci Transl Med 2018; 9:9/375/eaah6510. [PMID: 28148846 DOI: 10.1126/scitranslmed.aah6510] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 07/26/2016] [Accepted: 10/05/2016] [Indexed: 12/13/2022]
Abstract
Engineered neural stem cells (NSCs) are a promising approach to treating glioblastoma (GBM). The ideal NSC drug carrier for clinical use should be easily isolated and autologous to avoid immune rejection. We transdifferentiated (TD) human fibroblasts into tumor-homing early-stage induced NSCs (h-iNSCTE), engineered them to express optical reporters and different therapeutic gene products, and assessed the tumor-homing migration and therapeutic efficacy of cytotoxic h-iNSCTE in patient-derived GBM models of surgical and nonsurgical disease. Molecular and functional analysis revealed that our single-factor SOX2 TD strategy converted human skin fibroblasts into h-iNSCTE that were nestin+ and expressed pathways associated with tumor-homing migration in 4 days. Time-lapse motion analysis showed that h-iNSCTE rapidly migrated to human GBM cells and penetrated human GBM spheroids, a process inhibited by blockade of CXCR4. Serial imaging showed that h-iNSCTE delivery of the proapoptotic agent tumor necrosis factor-α-related apoptosis-inducing ligand (TRAIL) reduced the size of solid human GBM xenografts 250-fold in 3 weeks and prolonged median survival from 22 to 49 days. Additionally, h-iNSCTE thymidine kinase/ganciclovir enzyme/prodrug therapy (h-iNSCTE-TK) reduced the size of patient-derived GBM xenografts 20-fold and extended survival from 32 to 62 days. Mimicking clinical NSC therapy, h-iNSCTE-TK therapy delivered into the postoperative surgical resection cavity delayed the regrowth of residual GBMs threefold and prolonged survival from 46 to 60 days. These results suggest that TD of human skin into h-iNSCTE is a platform for creating tumor-homing cytotoxic cell therapies for cancer, where the potential to avoid carrier rejection could maximize treatment durability in human trials.
Collapse
Affiliation(s)
- Juli R Bagó
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Onyi Okolie
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Raluca Dumitru
- UNC Human Pluripotent Stem Cell Core Facility, Department of Genetics, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew G Ewend
- Department of Neurosurgery, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ryan Vander Werff
- Department of Cellular and Physiological Sciences, Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - T Michael Underhill
- Department of Cellular and Physiological Sciences, Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Ralf S Schmid
- Division of Neuropathology and Department of Pathology and Laboratory Medicine, Department of Neurology and Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - C Ryan Miller
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Division of Neuropathology and Department of Pathology and Laboratory Medicine, Department of Neurology and Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shawn D Hingtgen
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. .,Department of Neurosurgery, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
28
|
Delivery of Cytotoxic Mesenchymal Stem Cells with Biodegradable Scaffolds for Treatment of Postoperative Brain Cancer. Methods Mol Biol 2018; 1831:49-58. [PMID: 30051424 DOI: 10.1007/978-1-4939-8661-3_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Engineered stem cells have recently entered clinical trials as therapeutic agents for treating glioblastoma foci that remain after primary brain tumor resection. However, efficient delivery of anti-cancer mesenchymal stem cells (MSCs) into the resection cavity remains a potential obstacle to therapeutic efficacy in humans. Direct injection quickly leads to significant stem cell loss and poor tumor killing. Recent reports have shown that biodegradable scaffolds improve MSC persistence and restore therapeutic potential. Here, we describe a method for the delivery of therapeutic MSCs on biodegradable fibrin scaffolds into the resection cavity to treat postoperative brain cancer.
Collapse
|
29
|
|
30
|
Gulati K, Meher MK, Poluri KM. Glycosaminoglycan-based resorbable polymer composites in tissue refurbishment. Regen Med 2017. [DOI: 10.2217/rme-2017-0012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Regeneration of tissue structure with the aid of bioactive polymer matrices/composites and scaffolds for respective applications is one of the emerging areas of biomedical engineering. Recent advances in conjugated glycosaminoglycan (GAG) hybrids using natural and synthetic polymers have opened new avenues for producing a wide variety of resorbable polymer matrices. These hybrid scaffolds are low-immunogenic, highly biocompatible and biodegradable with incredible mechanical and tensile properties. GAG-based resorbable polymeric matrices are being exploited in migration of stem cells, cartilage and bone replacement/regeneration and production of scaffolds for various tissue engineering applications. In the current review, we will discuss the role of GAG-based resorbable polymer matrices in the field of regenerative medicine.
Collapse
Affiliation(s)
- Khushboo Gulati
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Mukesh Kumar Meher
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|
31
|
A bioorthogonal nanosystem for imaging and in vivo tumor inhibition. Biomaterials 2017; 138:57-68. [PMID: 28554008 DOI: 10.1016/j.biomaterials.2017.05.036] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/16/2017] [Accepted: 05/21/2017] [Indexed: 01/08/2023]
Abstract
Bioorthogonal bond-cleavage reactions have emerged as promising tools for manipulating biological processes, still the therapeutic effect of these reactions in vivo needs to be explored. Herein a bioorthogonal-activated prodrug has been developed for bioimaging and therapy, which is composed of a Pd-mediated cleavable propargyl, a coumarin fluorophore and a potent anticancer drug. In vitro investigations show that, the presence of a Pd complex induces the cleavage of propargyl and subsequently trigger the cascade of reactions, thereby activating the coumarin fluorophore for imaging and releasing the anticancer drug for therapy. Both the prodrug and Pd complex were then separately encapsulated into phospholipid liposomes to form a two-component bioorthogonal nanosystem. The lyposomal nanosystem can be readily internalized by HeLa cells and displays strong intracellular fluorescence under one- or two-photon excitation, indicating the release of the active drug in cells as a result of the Pd-mediated bioorthogonal bond-cleavage reaction. More importantly, the nanosystem shows considerable high activity and exerts efficient inhibition towards tumor growth in a mouse model. This work demonstrates that, if properly formulated, a bioorthogonal system can perform well in vivo. This strategy may offer a new approach for designing bioorthogonal prodrugs with imaging and therapeutic capability.
Collapse
|
32
|
Bagó JR, Pegna GJ, Okolie O, Mohiti-Asli M, Loboa EG, Hingtgen SD. Electrospun nanofibrous scaffolds increase the efficacy of stem cell-mediated therapy of surgically resected glioblastoma. Biomaterials 2016; 90:116-25. [PMID: 27016620 DOI: 10.1016/j.biomaterials.2016.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 03/02/2016] [Accepted: 03/06/2016] [Indexed: 12/16/2022]
Abstract
Engineered stem cell (SC)-based therapy holds enormous promise for treating the incurable brain cancer glioblastoma (GBM). Retaining the cytotoxic SCs in the surgical cavity after GBM resection is one of the greatest challenges to this approach. Here, we describe a biocompatible electrospun nanofibrous scaffold (bENS) implant capable of delivering and retaining tumor-homing cytotoxic stem cells that suppress recurrence of post-surgical GBM. As a new approach to GBM therapy, we created poly(l-lactic acid) (PLA) bENS bearing drug-releasing human mesenchymal stem cells (hMSCs). We discovered that bENS-based implant increased hMSC retention in the surgical cavity 5-fold and prolonged persistence 3-fold compared to standard direct injection using our mouse model of GBM surgical resection/recurrence. Time-lapse imaging showed cytotoxic hMSC/bENS treatment killed co-cultured human GBM cells, and allowed hMSCs to rapidly migrate off the scaffolds as they homed to GBMs. In vivo, bENS loaded with hMSCs releasing the anti-tumor protein TRAIL (bENS(sTR)) reduced the volume of established GBM xenografts 3-fold. Mimicking clinical GBM patient therapy, lining the post-operative GBM surgical cavity with bENS(sTR) implants inhibited the re-growth of residual GBM foci 2.3-fold and prolonged post-surgical median survival from 13.5 to 31 days in mice. These results suggest that nanofibrous-based SC therapies could be an innovative new approach to improve the outcomes of patients suffering from terminal brain cancer.
Collapse
Affiliation(s)
- Juli R Bagó
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Guillaume J Pegna
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Onyi Okolie
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mahsa Mohiti-Asli
- Joint Department of Biomedical Engineering at University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
| | - Elizabeth G Loboa
- Joint Department of Biomedical Engineering at University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA; College of Engineering, University of Missouri, Columbia, MO, 65211, USA
| | - Shawn D Hingtgen
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|