1
|
Bardhan M, Muneer MA, Khare A, Minesh Shah R, Kaur A, Vasipalli SS, Suresh V, Podder V, Ahluwalia M, Odia Y, Chen Z. Advances in stem cell-based therapeutic transfers for glioblastoma treatment. Expert Rev Neurother 2025:1-17. [PMID: 40245098 DOI: 10.1080/14737175.2025.2490543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Glioblastoma (GBM), a highly malignant brain tumor, has a poor prognosis despite standard treatments like surgery, chemotherapy, and radiation. Glioblastoma stem cells (GSCs) play a critical role in recurrence and therapy resistance. Stem cell-based therapies have emerged as innovative approaches, leveraging the tumor-targeting abilities of stem cells to deliver treatments directly to GBM. AREAS COVERED This review focuses on using intact stem cells or subtypes for GBM therapy, excluding antigenic characteristics. The stem cell-based therapies explored include neural, mesenchymal, glioblastoma, hematopoietic and adipose-derived stem cells that have been investigated in both clinical and preclinical settings. A systematic search in PubMed, EMBASE, ClinicalTrials.gov, and Scopus had identified research up until January 2024. Key mechanisms reviewed include immune modulation, angiogenesis inhibition, and apoptosis induction. Discussion of completed and ongoing trials include emphasis on safety, efficacy, challenges, and study design limitations. EXPERT OPINION Stem cell-based therapies hold promise for treating GBM by targeting GSCs and improving treatment outcomes. Despite some potential advantages, challenges such as tumorigenesis risks, delivery complexities, and sustained therapeutic effects persist. Future research should prioritize optimizing stem cell modifications, combining them with current treatments, and conducting large-scale trials to ensure safety and efficacy. Integrating stem cell therapies into GBM treatment could provide more effective and less invasive options for patients.
Collapse
Affiliation(s)
- Mainak Bardhan
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | | | - Abhinav Khare
- All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | | | - Anmol Kaur
- Lady Hardinge Medical College, New Delhi, India
| | - Sonit Sai Vasipalli
- Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Vinay Suresh
- King George's Medical University, Lucknow, India
| | - Vivek Podder
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Manmeet Ahluwalia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Yazmin Odia
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Zhijian Chen
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| |
Collapse
|
2
|
Peng Z, Kalim M, Lu Y. Improving systemic delivery of oncolytic virus by cellular carriers. Cancer Biol Med 2025; 21:j.issn.2095-3941.2024.0390. [PMID: 39831754 PMCID: PMC11745088 DOI: 10.20892/j.issn.2095-3941.2024.0390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025] Open
Abstract
Oncolytic virotherapy (OVT) is a promising option for cancer treatment. OVT involves selective oncolytic virus (OV) replication within cancer cells, which triggers anti-tumor responses and immunostimulation. Despite promising potential, OVT faces critical challenges, including insufficient tumor-specific targeting, which results in limited tumor penetration and variability in therapeutic efficacy. These challenges are particularly pronounced in solid tumors with complex microenvironments and heterogeneous vascularization. A comprehensive research program is currently underway to develop and refine innovative delivery methods to address these issues to enhance OVT precision and efficacy. A principal area of investigation is the utilization of cellular carriers to enhance the delivery and distribution of OVs within tumor microenvironments, thereby optimizing immune system activation and maximizing anti-tumor effects. This review offers a comprehensive overview of the current strategies that are being used to enhance the delivery of OVs via cellular carriers with the goal of improving the clinical impact of OVT in cancer therapy.
Collapse
Affiliation(s)
- Ziyi Peng
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|
3
|
Liu C, Gao J, Cheng Y, Zhang S, Fu C. Homologous-adhering/targeting cell membrane- and cell-mediated delivery systems: a cancer-catch-cancer strategy in cancer therapy. Regen Biomater 2024; 12:rbae135. [PMID: 39811105 PMCID: PMC11729729 DOI: 10.1093/rb/rbae135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/09/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Low tumor enrichment remains a serious and urgent problem for drug delivery in cancer therapy. Accurate targeting of tumor sites is still a critical aim in cancer therapy. Though there have been a variety of delivery strategies to improve the tumor targeting and enrichment, biological barriers still cause most delivered guests to fail or be excreted before they work. Recently, cell membrane-based systems have attracted a huge amount of attention due to their advantages such as easy access, good biocompatibility and immune escape, which contribute to their biomimetic structures and specific surface proteins. Furthermore, cancer cell membrane-based delivery systems are referred to as homologous-targeting function in which they exhibit significantly high adhesion and internalization to homologous-type tumor sites or cells even though the exact mechanism is not entirely revealed. Here, we summarize the sources and characterizations of cancer cell membrane systems, including reconstructed single or hybrid membrane-based nano-/microcarriers, as well as engineered cancer cells. Additionally, advanced applications of these cancer cell membrane systems in cancer therapy are categorized and summarized according to the components of membranes. The potential factors related to homologous targeting of cancer cell membrane-based systems are also discussed. By discussing the applications, challenges and opportunities, we expect the cancer cell membrane-based homologous-targeting systems to have a far-reaching development in preclinic or clinics.
Collapse
Affiliation(s)
- Chenguang Liu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Jingjie Gao
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Yuying Cheng
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Shanshan Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Caiyun Fu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| |
Collapse
|
4
|
Rodríguez-Mendoza B, Figueroa-González A, Cano-Herrera G, Gutiérrez-Rosas LE, Romero-Torres CI, Victoria-García LO, González-Castillo P, Guerrero-Cázares H, Ibarra A. [Glioblastoma and its interaction with neurogenesis]. Rev Neurol 2024; 79:279-287. [PMID: 39540380 PMCID: PMC11605900 DOI: 10.33588/rn.7910.2024226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive malignant primary tumor of the central nervous system in adults, with an incidence of 3.23 per 100,000 people. Despite the existence of various therapeutic approaches, the absence of a cure and the unfavorable prognosis persist for this neoplasm, with a median survival of approximately 8-15 months and a 5-year survival rate of 6.9%. In this review, we address the epidemiology, histopathology, molecular characteristics, and treatment of GBM. We highlight the relationship of GBM with the microenvironment in the lateral ventricle wall and the cerebrospinal fluid. The location of GBM in this region results in more aggressive tumors and shorter life expectancy for patients. Understanding the malignancy mechanisms that hinder remission, treatment, and positive prognosis opens the possibility of improving diagnostic and therapeutic interventions against GBM.
Collapse
Affiliation(s)
- Brenda Rodríguez-Mendoza
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Aura Figueroa-González
- Neurosurgery Department. Mayo Clinic Jacksonville. Jacksonville, Estados UnidosMayo Clinic JacksonvilleMayo Clinic JacksonvilleJacksonvilleEstados Unidos
| | - Gabriela Cano-Herrera
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Luis E. Gutiérrez-Rosas
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Carlos I. Romero-Torres
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Luis O. Victoria-García
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Paola González-Castillo
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Hugo Guerrero-Cázares
- Neurosurgery Department. Mayo Clinic Jacksonville. Jacksonville, Estados UnidosMayo Clinic JacksonvilleMayo Clinic JacksonvilleJacksonvilleEstados Unidos
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
- Secretaría de la Defensa Nacional. Escuela Militar de Graduados en Sanidad. Ciudad de México, MéxicoEscuela Militar de Graduados en SanidadEscuela Militar de Graduados en SanidadCiudad de MéxicoMéxico
| |
Collapse
|
5
|
Kass L, Thang M, Zhang Y, DeVane C, Logan J, Tessema A, Perry J, Hingtgen S. Development of a biocompatible 3D hydrogel scaffold using continuous liquid interface production for the delivery of cell therapies to treat recurrent glioblastoma. Bioeng Transl Med 2024; 9:e10676. [PMID: 39545092 PMCID: PMC11558199 DOI: 10.1002/btm2.10676] [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: 01/17/2024] [Revised: 03/21/2024] [Accepted: 04/18/2024] [Indexed: 11/17/2024] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor diagnosed in adults, carrying with it an extremely poor prognosis and limited options for effective treatment. Various cell therapies have emerged as promising candidates for GBM treatment but fail in the clinic due to poor tumor trafficking, poor transplantation efficiency, and high systemic toxicity. In this study, we design, characterize, and test a 3D-printed cell delivery platform that can enhance the survival of therapeutic cells implanted in the GBM resection cavity. Using continuous liquid interface production (CLIP) to generate a biocompatible 3D hydrogel, we demonstrate that we can effectively seed neural stem cells (NSCs) onto the surface of the hydrogel, and that the cells can proliferate to high densities when cultured for 14 days in vitro. We show that NSCs seeded on CLIP scaffolds persist longer than freely injected cells in vivo, proliferating to 20% higher than their original density in 6 days after implantation. Finally, we demonstrate that therapeutic fibroblasts seeded on CLIP more effectively suppress tumor growth and extend survival in a mouse model of LN229 GBM resection compared to the scaffold or therapeutic cells alone. These promising results demonstrate the potential to leverage CLIP to design hydrogels with various features to control the delivery of different types of cell therapies. Future work will include a more thorough evaluation of the immunological response to the material and improvement of the printing resolution for biocompatible aqueous resins.
Collapse
Affiliation(s)
- Lauren Kass
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Morrent Thang
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Yu Zhang
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Cathleen DeVane
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Julia Logan
- Department of Chemistry, UNC College of Arts and SciencesThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Addis Tessema
- Department of Chemistry, UNC College of Arts and SciencesThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Jillian Perry
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Center for Nanotechnology in Drug Delivery, Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Shawn Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of PharmacyThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterThe University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
6
|
Li Y, Fan Y, Xie Y, Li L, Li J, Liu J, Jin Z, Xue H, Wang Z. Genome-wide RNA-Seq identifies TP53-mediated embryonic stem cells inhibiting tumor invasion and metastasis. Stem Cell Res Ther 2024; 15:369. [PMID: 39415294 PMCID: PMC11483973 DOI: 10.1186/s13287-024-04000-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/14/2024] [Indexed: 10/18/2024] Open
Abstract
The discovery of embryonic stem cell (ESC) mediating tumoricidal activity revealed the intimate relationship between ESCs and tumor cells, but the functional role of ESCs in tumor progression is poorly understood. To further investigate tumor cell and ESC interactions, we co-cultured mouse ESCs with mouse pancreatic cancer Pan02 cells or mouse melanoma B16-F10 cells in Transwell, and found that tumor cell invasion was significantly inhibited by ESCs. Application of ESCs to tumor-bearing mice resulted in significant inhibition of tumor metastasis in vivo. RNA-Seq analyses of tumor cell and ESC co-cultures identified TP53 and related signalling as major pathways involved in ESC-mediated inhibition of tumor cell invasion and metastasis, which indicated the potential clinical application of ESCs to treat cancer.
Collapse
Affiliation(s)
- Yatong Li
- Department of Radiology, Peking Union Medical College Hospital, Beijing, 100730, China.
- State Key Laboratory of Medical Molecular Biology, Department of Pathology and Pathophysiology, Institute of Basic Medical Sciences, Center of Molecular Pathology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China.
| | - Yongna Fan
- State Key Laboratory of Medical Molecular Biology, Department of Pathology and Pathophysiology, Institute of Basic Medical Sciences, Center of Molecular Pathology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Yunyi Xie
- State Key Laboratory of Medical Molecular Biology, Department of Pathology and Pathophysiology, Institute of Basic Medical Sciences, Center of Molecular Pathology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Limin Li
- State Key Laboratory of Medical Molecular Biology, Department of Pathology and Pathophysiology, Institute of Basic Medical Sciences, Center of Molecular Pathology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100005, China
| | - Juan Li
- Department of Radiology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Jingyi Liu
- Department of Radiology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Zhiwei Wang
- Department of Radiology, Peking Union Medical College Hospital, Beijing, 100730, China.
| |
Collapse
|
7
|
Zeng X, Ropper AE, Aljuboori Z, Yu D, Teng TW, Kabatas S, Usuga E, Anderson JE, Teng YD. Concurrent Oncolysis and Neurolesion Repair by Dual Gene-Engineered hNSCs in an Experimental Model of Intraspinal Cord Glioblastoma. Cells 2024; 13:1522. [PMID: 39329707 PMCID: PMC11429792 DOI: 10.3390/cells13181522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 09/28/2024] Open
Abstract
Intramedullary spinal cord glioblastoma (ISCG) is lethal due to lack of effective treatment. We previously established a rat C6-ISCG model and the antitumor effect of F3.CD-TK, an hNSC line expressing CD and TK, via producing cytocidal 5FU and GCV-TP. However, the neurotherapeutic potential of this hNSC approach has remained uninvestigated. Here for the first time, cultured F3.CD-TK cells were found to have a markedly higher oncolytic effect, which was GJIC-dependent, and BDNF expression but less VEGF secretion than F3.CD. In Rowett athymic rats, F3.CD-TK (1.5 × 106 cells/10 µL × 2), injected near C6-ISCG (G55 seeding 7 days earlier: 10 K/each) and followed by q.d. (×5/each repeat; i.p.) of 5FC (500 mg/kg/5 mL/day) and GCV (25 mg/kg/1 mL/day), robustly mitigated cardiorespiratory, locomotor, and sensory deficits to improve neurofunction and overall survival compared to animals receiving either F3.CD or F3.CD-TK+F3.CD debris formula. The F3.CD-TK regimen exerted greater tumor penetration and neural inflammation/immune modulation, reshaped C6-ISCG topology to increase the tumor's surface area/volume ratio to spare/repair host axons (e.g., vGlut1+ neurites), and had higher post-prodrug donor self-clearance. The multimodal data and mechanistic leads from this proof-of-principle study suggest that the overall stronger anti-ISCG benefit of our hNSC-based GDEPT is derived from its concurrent oncolytic and neurotherapeutic effects.
Collapse
Affiliation(s)
- Xiang Zeng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Alexander E. Ropper
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Zaid Aljuboori
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Dou Yu
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | | | - Serdar Kabatas
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Esteban Usuga
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Jamie E. Anderson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Yang D. Teng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
8
|
Oishi T, Koizumi S, Kurozumi K. Mesenchymal stem cells as therapeutic vehicles for glioma. Cancer Gene Ther 2024; 31:1306-1314. [PMID: 38654128 DOI: 10.1038/s41417-024-00775-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Glioma is a disease with a poor prognosis despite the availability of multimodality treatments, and the development of novel therapies is urgently needed. Challenges in glioma treatment include the difficulty for drugs to cross the blood-brain barrier when administered systemically and poor drug diffusion when administered locally. Mesenchymal stem cells exhibit advantages for glioma therapy because of their ability to pass through the blood-brain barrier and migrate to tumor cells and their tolerance to the immune system. Therefore, mesenchymal stem cells have been explored as vehicles for various therapeutic agents for glioma treatment. Mesenchymal stem cells loaded with chemotherapeutic drugs show improved penetration and tumor accumulation. For gene therapy, mesenchymal stem cells can be used as vehicles for suicide genes, the so-called gene-directed enzyme prodrug therapy. Mesenchymal stem cell-based oncolytic viral therapies have been attempted in recent years to enhance the efficacy of infection against the tumor, viral replication, and distribution of viral particles. Many uncertainties remain regarding the function and behavior of mesenchymal stem cells in gliomas. However, strategies to increase mesenchymal stem cell migration to gliomas may improve the delivery of therapeutic agents and enhance their anti-tumor effects, representing promising potential for patient treatment.
Collapse
Affiliation(s)
- Tomoya Oishi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinichiro Koizumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiko Kurozumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
9
|
Manchanda AS, Rai HK, Kaur M, Arora P. Cancer stem cells targeted therapy: A changing concept in head and neck squamous cell carcinoma. J Oral Maxillofac Pathol 2024; 28:455-463. [PMID: 39670113 PMCID: PMC11633930 DOI: 10.4103/jomfp.jomfp_248_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 12/14/2024] Open
Abstract
Identification of cancer stem cells (CSCs), their multilineage potential, and their ability of self-renewal have revolutionised the current concepts of cancer treatment. The suspected role of CSCs in cancer initiation, progression and relapse with the observed resistance to conventional cancer treatments has led to the development of more specific and targeted therapies. Identification of the properties of stem cells (SCs) and their potential for localisation in cancer has made targeted anti-cancer treatment possible by incorporating some modifications into these SCs. The same concept has been applied to the treatment strategy for head and neck squamous cell carcinoma (HNSCC) to control the relapse and improve the mortality rates in patients. This review aims to discuss the role of CSCs in the course and relapse of HNSCC, various surface markers for their identification and SC-targeted therapy options for the treatment of HNSCC, with a highlight on the advantages, shortcomings, opportunities and challenges to SC therapy in head and neck squamous cell carcinoma, treatment and scope for future research.
Collapse
Affiliation(s)
- Adesh S. Manchanda
- Department of Oral and Maxillofacial Pathology, Sri Guru Ram Das Institute of Dental Sciences and Research, Amritsar, Punjab, India
| | - Harmandeep K. Rai
- Department of Dentistry, Sri Guru Ram Das Institute of Dental Sciences and Research, Amritsar, Punjab, India
| | - Manvir Kaur
- Department of Dentistry, Sri Guru Ram Das Institute of Dental Sciences and Research, Amritsar, Punjab, India
| | - Paras Arora
- Department of Dentistry, Sri Guru Ram Das Institute of Dental Sciences and Research, Amritsar, Punjab, India
| |
Collapse
|
10
|
Kumawat C, Takahashi T, Date I, Tomita Y, Tanaka M, Arataki S, Komatsubara T, Flores AOP, Yu D, Jain M. State-of-the-Art and New Treatment Approaches for Spinal Cord Tumors. Cancers (Basel) 2024; 16:2360. [PMID: 39001422 PMCID: PMC11240441 DOI: 10.3390/cancers16132360] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Spinal cord tumors, though rare, present formidable challenges in clinical management due to their intricate nature. Traditional treatment modalities like surgery, radiation therapy, and chemotherapy have been the mainstay for managing these tumors. However, despite significant advancements, challenges persist, including the limitations of surgical resection and the potential side effects associated with radiation therapy. In response to these limitations, a wave of innovative approaches is reshaping the treatment landscape for spinal cord tumors. Advancements in gene therapy, immunotherapy, and targeted therapy are offering groundbreaking possibilities. Gene therapy holds the potential to modify the genes responsible for tumor growth, while immunotherapy harnesses the body's own immune system to fight cancer cells. Targeted therapy aims to strike a specific vulnerability within the tumor cells, offering a more precise and potentially less toxic approach. Additionally, novel surgical adjuncts are being explored to improve visualization and minimize damage to surrounding healthy tissue during tumor removal. These developments pave the way for a future of personalized medicine for spinal cord tumors. By delving deeper into the molecular makeup of individual tumors, doctors can tailor treatment strategies to target specific mutations and vulnerabilities. This personalized approach offers the potential for more effective interventions with fewer side effects, ultimately leading to improved patient outcomes and a better quality of life. This evolving landscape of spinal cord tumor management signifies the crucial integration of established and innovative strategies to create a brighter future for patients battling this complex condition.
Collapse
Affiliation(s)
- Chetan Kumawat
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
- Department of Orthopedic Surgery, Sir Ganga Ram Hospital, Rajinder Nagar, New Delhi 110060, India
| | - Toshiyuki Takahashi
- Spinal Disorder Center, Fujieda Heisei Memorial Hospital, 123-1 Mizuue Fujieda, Shizuoka 426-8662, Japan
| | - Isao Date
- Department of Neurosurgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Yousuke Tomita
- Department of Neurosurgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Masato Tanaka
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Shinya Arataki
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Tadashi Komatsubara
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Angel O P Flores
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Dongwoo Yu
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| | - Mukul Jain
- Department of Orthopedic Surgery, Okayama Rosai Hospital, 1-10-25 Chikkomidorimachi, Minami Ward Okayama, Okayama 702-8055, Japan
| |
Collapse
|
11
|
Rayati M, Mansouri V, Ahmadbeigi N. Gene therapy in glioblastoma multiforme: Can it be a role changer? Heliyon 2024; 10:e27087. [PMID: 38439834 PMCID: PMC10909773 DOI: 10.1016/j.heliyon.2024.e27087] [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: 09/26/2023] [Revised: 02/07/2024] [Accepted: 02/23/2024] [Indexed: 03/06/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal cancers with a poor prognosis. Over the past century since its initial discovery and medical description, the development of effective treatments for this condition has seen limited progress. Despite numerous efforts, only a handful of drugs have gained approval for its treatment. However, these treatments have not yielded substantial improvements in both overall survival and progression-free survival rates. One reason for this is its unique features such as heterogeneity and difficulty of drug delivery because of two formidable barriers, namely the blood-brain barrier and the tumor-blood barrier. Over the past few years, significant developments in therapeutic approaches have given rise to promising novel and advanced therapies. Target-specific therapies, such as monoclonal antibodies (mAbs) and small molecules, stand as two important examples; however, they have not yielded a significant improvement in survival among GBM patients. Gene therapy, a relatively nascent advanced approach, holds promise as a potential treatment for cancer, particularly GBM. It possesses the potential to address the limitations of previous treatments and even newer advanced therapies like mAbs, owing to its distinct properties. This review aims to elucidate the current status and advancements in gene therapy for GBM treatment, while also presenting its future prospects.
Collapse
Affiliation(s)
- Mohammad Rayati
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Naser Ahmadbeigi
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
El‐Ayoubi A, Arakelyan A, Klawitter M, Merk L, Hakobyan S, Gonzalez‐Menendez I, Quintanilla Fend L, Holm PS, Mikulits W, Schwab M, Danielyan L, Naumann U. Development of an optimized, non-stem cell line for intranasal delivery of therapeutic cargo to the central nervous system. Mol Oncol 2024; 18:528-546. [PMID: 38115217 PMCID: PMC10920084 DOI: 10.1002/1878-0261.13569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/23/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Neural stem cells (NSCs) are considered to be valuable candidates for delivering a variety of anti-cancer agents, including oncolytic viruses, to brain tumors. However, owing to the previously reported tumorigenic potential of NSC cell lines after intranasal administration (INA), here we identified the human hepatic stellate cell line LX-2 as a cell type capable of longer resistance to replication of oncolytic adenoviruses (OAVs) as a therapeutic cargo, and that is non-tumorigenic after INA. Our data show that LX-2 cells can longer withstand the OAV XVir-N-31 replication and oncolysis than NSCs. By selecting the highly migratory cell population out of LX-2, an offspring cell line with a higher and more stable capability to migrate was generated. Additionally, as a safety backup, we applied genomic herpes simplex virus thymidine kinase (HSV-TK) integration into LX-2, leading to high vulnerability to ganciclovir (GCV). Histopathological analyses confirmed the absence of neoplasia in the respiratory tracts and brains of immuno-compromised mice 3 months after INA of LX-2 cells. Our data suggest that LX-2 is a novel, robust, and safe cell line for delivering anti-cancer and other therapeutic agents to the brain.
Collapse
Affiliation(s)
- Ali El‐Ayoubi
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center NeurologyUniversity Hospital of TübingenGermany
| | - Arsen Arakelyan
- Research Group of BioinformaticsInstitute of Molecular Biology NAS RAYerevanArmenia
| | - Moritz Klawitter
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center NeurologyUniversity Hospital of TübingenGermany
| | - Luisa Merk
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center NeurologyUniversity Hospital of TübingenGermany
| | - Siras Hakobyan
- Research Group of BioinformaticsInstitute of Molecular Biology NAS RAYerevanArmenia
- Armenian Institute of BioinformaticsYerevanArmenia
| | - Irene Gonzalez‐Menendez
- Institute for Pathology, Department of General and Molecular PathologyUniversity Hospital TübingenGermany
- Cluster of Excellence iFIT (EXC 2180) "Image‐Guided and Functionally Instructed Tumor Therapies"Eberhard Karls University of TübingenGermany
| | - Leticia Quintanilla Fend
- Institute for Pathology, Department of General and Molecular PathologyUniversity Hospital TübingenGermany
- Cluster of Excellence iFIT (EXC 2180) "Image‐Guided and Functionally Instructed Tumor Therapies"Eberhard Karls University of TübingenGermany
| | - Per Sonne Holm
- Department of Urology, Klinikum rechts der IsarTechnical University of MunichGermany
- Department of Oral and Maxillofacial SurgeryMedical University InnsbruckAustria
- XVir Therapeutics GmbHMunichGermany
| | - Wolfgang Mikulits
- Center for Cancer Research, Comprehensive Cancer CenterMedical University of ViennaAustria
| | - Matthias Schwab
- Cluster of Excellence iFIT (EXC 2180) "Image‐Guided and Functionally Instructed Tumor Therapies"Eberhard Karls University of TübingenGermany
- Dr. Margarete Fischer‐Bosch Institute of Clinical PharmacologyStuttgartGermany
- Department of Pharmacy and BiochemistryUniversity of TübingenGermany
- Department of Clinical PharmacologyUniversity Hospital TübingenGermany
- Neuroscience Laboratory and Departments of Biochemistry and Clinical PharmacologyYerevan State Medical UniversityArmenia
| | - Lusine Danielyan
- Department of Pharmacy and BiochemistryUniversity of TübingenGermany
- Department of Clinical PharmacologyUniversity Hospital TübingenGermany
- Neuroscience Laboratory and Departments of Biochemistry and Clinical PharmacologyYerevan State Medical UniversityArmenia
| | - Ulrike Naumann
- Molecular Neurooncology, Department of Vascular Neurology, Hertie Institute for Clinical Brain Research and Center NeurologyUniversity Hospital of TübingenGermany
- Gene and RNA Therapy Center (GRTC)Faculty of Medicine University TübingenGermany
| |
Collapse
|
13
|
Ballestín A, Armocida D, Ribecco V, Seano G. Peritumoral brain zone in glioblastoma: biological, clinical and mechanical features. Front Immunol 2024; 15:1347877. [PMID: 38487525 PMCID: PMC10937439 DOI: 10.3389/fimmu.2024.1347877] [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: 12/01/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Glioblastoma is a highly aggressive and invasive tumor that affects the central nervous system (CNS). With a five-year survival rate of only 6.9% and a median survival time of eight months, it has the lowest survival rate among CNS tumors. Its treatment consists of surgical resection, subsequent fractionated radiotherapy and concomitant and adjuvant chemotherapy with temozolomide. Despite the implementation of clinical interventions, recurrence is a common occurrence, with over 80% of cases arising at the edge of the resection cavity a few months after treatment. The high recurrence rate and location of glioblastoma indicate the need for a better understanding of the peritumor brain zone (PBZ). In this review, we first describe the main radiological, cellular, molecular and biomechanical tissue features of PBZ; and subsequently, we discuss its current clinical management, potential local therapeutic approaches and future prospects.
Collapse
Affiliation(s)
- Alberto Ballestín
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Daniele Armocida
- Human Neurosciences Department, Neurosurgery Division, Sapienza University, Rome, Italy
| | - Valentino Ribecco
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| | - Giorgio Seano
- Tumor Microenvironment Laboratory, UMR3347 CNRS/U1021 INSERM, Institut Curie, Orsay, France
| |
Collapse
|
14
|
Tolboom N, Verger A, Albert NL, Fraioli F, Guedj E, Traub-Weidinger T, Morbelli S, Herrmann K, Zucchetta P, Plasschaert SLA, Yakushev I, Weller M, Glas M, Preusser M, Cecchin D, Barthel H, Van Weehaeghe D. Theranostics in Neurooncology: Heading Toward New Horizons. J Nucl Med 2024; 65:167-173. [PMID: 38071569 DOI: 10.2967/jnumed.123.266205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/23/2023] [Indexed: 02/03/2024] Open
Abstract
Therapeutic approaches to brain tumors remain a challenge, with considerable limitations regarding delivery of drugs. There has been renewed and increasing interest in translating the popular theranostic approach well known from prostate and neuroendocrine cancer to neurooncology. Although far from perfect, some of these approaches show encouraging preliminary results, such as for meningioma and leptomeningeal spread of certain pediatric brain tumors. In brain metastases and gliomas, clinical results have failed to impress. Perspectives on these theranostic approaches regarding meningiomas, brain metastases, gliomas, and common pediatric brain tumors will be discussed. For each tumor entity, the general context, an overview of the literature, and future perspectives will be provided. Ongoing studies will be discussed in the supplemental materials. As most theranostic agents are unlikely to cross the blood-brain barrier, the delivery of these agents will be dependent on the successful development and clinical implementation of techniques enhancing permeability and retention. Moreover, the international community should strive toward sufficiently large and randomized studies to generate high-level evidence on theranostic approaches with radioligand therapies for central nervous system tumors.
Collapse
Affiliation(s)
- Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Antoine Verger
- IADI, INSERM, UMR 1254, Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU-Nancy, Université de Lorraine, Nancy, France
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital of Munich, Munich, Germany
| | - Francesco Fraioli
- Institute of Nuclear Medicine, University College London, London, United Kingdom
| | - Eric Guedj
- Département de Médecine Nucléaire, Hôpital de la Timone, CERIMED, Institut Fresnel, Aix Marseille University, APHM, CNRS, Centrale Marseille, Marseille, France
| | - Tatjana Traub-Weidinger
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa Italy
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium-University Hospital Essen, Essen, Germany
| | - Pietro Zucchetta
- Department of Nuclear Medicine, University Hospital of Padova, Padova, Italy
| | | | - Igor Yakushev
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich and Munich Center for Neurosciences-Brain and Mind, Munich, Germany
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Medicine Essen, University Duisburg-Essen and German Cancer Consortium, Essen, Germany
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine-DIMED, University Hospital of Padua, Padua, Italy
| | - Henryk Barthel
- Department of Nuclear Medicine, Leipzig University Medical Centre, Leipzig, Germany; and
| | | |
Collapse
|
15
|
Kosianova А, Pak O, Bryukhovetskiy I. Regulation of cancer stem cells and immunotherapy of glioblastoma (Review). Biomed Rep 2024; 20:24. [PMID: 38170016 PMCID: PMC10758921 DOI: 10.3892/br.2023.1712] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Glioblastoma (GB) is one of the most adverse diagnoses in oncology. Complex current treatment results in a median survival of 15 months. Resistance to treatment is associated with the presence of cancer stem cells (CSCs). The present review aimed to analyze the mechanisms of CSC plasticity, showing the particular role of β-catenin in regulating vital functions of CSCs, and to describe the molecular mechanisms of Wnt-independent increase of β-catenin levels, which is influenced by the local microenvironment of CSCs. The present review also analyzed the reasons for the low effectiveness of using medication in the regulation of CSCs, and proposed the development of immunotherapy scenarios with tumor cell vaccines, containing heterogenous cancer cells able of producing a multidirectional antineoplastic immune response. Additionally, the possibility of managing lymphopenia by transplanting hematopoietic stem cells from a healthy sibling and using clofazimine or other repurposed drugs that reduce β-catenin concentration in CSCs was discussed in the present study.
Collapse
Affiliation(s)
- Аleksandra Kosianova
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Oleg Pak
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Igor Bryukhovetskiy
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| |
Collapse
|
16
|
Varela ML, Comba A, Faisal SM, Argento A, Peña Aguelo JA, Candolfi M, Castro MG, Lowenstein PR. Cell and gene therapy in neuro-oncology. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:297-315. [PMID: 39341660 PMCID: PMC11441620 DOI: 10.1016/b978-0-323-90120-8.00009-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The majority of primary brain tumors are gliomas, among which glioblastoma multiforme (GBM) is the most common malignant brain tumor in adults. GBM has a median survival of 18-24 months, and despite extensive research it remains incurable, thus novel therapies are urgently needed. The current standard of care is a combination of surgery, radiation, and chemotherapy, but still remains ineffective due to the invasive nature and high recurrence of gliomas. Gene therapy is a versatile treatment strategy investigated for multiple tumor types including GBM. In gene therapy, a variety of vectors are employed to deliver genes designed for different antitumoral effects. Also, over the past decades, stem cell biology has provided a new approach to cancer therapies. Stem cells can be used as regenerative medicine, therapeutic carriers, drug targeting, and generation of immune cells. Stem cell-based therapy allows targeted therapy that spares healthy brain tissue as well as establishes a long-term antitumor response by stimulating the immune system and delivering prodrug, metabolizing genes, or even oncolytic viruses. This chapter describes the latest developments and the current trends in gene and cell-based therapy against GBM from both preclinical and clinical perspectives, including different gene therapy delivery systems, molecular targets, and stem cell therapies.
Collapse
Affiliation(s)
- Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anna Argento
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Jorge A Peña Aguelo
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
17
|
Nasrolahi A, Shabani Z, Sadigh-Eteghad S, Salehi-Pourmehr H, Mahmoudi J. Stem Cell Therapy for the Treatment of Parkinson's Disease: What Promise Does it Hold? Curr Stem Cell Res Ther 2024; 19:185-199. [PMID: 36815638 DOI: 10.2174/1574888x18666230222144116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 02/24/2023]
Abstract
Parkinson's disease (PD) is a common, progressive neurodegenerative disorder characterized by substantia nigra dopamine cell death and a varied clinical picture that affects older people. Although more than two centuries have passed since the earliest attempts to find a cure for PD, it remains an unresolved problem. With this in mind, cell replacement therapy is a new strategy for treating PD. This novel approach aims to replace degenerated dopaminergic (DAergic) neurons with new ones or provide a new source of cells that can differentiate into DAergic neurons. Induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), neural stem cells (NSCs), and embryonic stem cells (ESCs) are among the cells considered for transplantation therapies. Recently disease-modifying strategies like cell replacement therapies combined with other therapeutic approaches, such as utilizing natural compounds or biomaterials, are proposed to modify the underlying neurodegeneration. In the present review, we discuss the current advances in cell replacement therapy for PD and summarize the existing experimental and clinical evidence supporting this approach.
Collapse
Affiliation(s)
- Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Cellular and Molecular Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Shabani
- Center for Cerebrovascular Research, University of California, San Francisco, California, USA
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hanieh Salehi-Pourmehr
- Research Center for Evidence-Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mahmoudi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Rosu A, Ghaemi B, Bulte JW, Shakeri-Zadeh A. Tumor-tropic Trojan horses: Using mesenchymal stem cells as cellular nanotheranostics. Theranostics 2024; 14:571-591. [PMID: 38169524 PMCID: PMC10758060 DOI: 10.7150/thno.90187] [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: 09/14/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024] Open
Abstract
Various classes of nanotheranostics have been developed for enhanced tumor imaging and therapy. However, key limitations for a successful use of nanotheranostics include their targeting specificity with limited off-site tissue accumulation as well as their distribution and prolonged retention throughout the entire tumor. Due to their inherent tumor-tropic properties, the use of mesenchymal stem cells (MSCs) as a "Trojan horse" has recently been proposed to deliver nanotheranostics more effectively. This review discusses the current status of "cellular nanotheranostics" for combined (multimodal) imaging and therapy in preclinical cancer models. Emphasis is placed on the limited knowledge of the signaling pathways and molecular mechanisms of MSC tumor-tropism, and how such information may be exploited to engineer MSCs in order to further improve tumor homing and nanotheranostic delivery using image-guided procedures.
Collapse
Affiliation(s)
| | | | | | - Ali Shakeri-Zadeh
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research and Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Xiao Y, Wu M, Xue C, Wang Y. Recent Advances in the Development of Membrane-derived Vesicles for Cancer Immunotherapy. Curr Drug Deliv 2024; 21:403-420. [PMID: 37143265 DOI: 10.2174/1567201820666230504120841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023]
Abstract
The surface proteins on cell membranes enable the cells to have different properties, such as high biocompatibility, surface modifiability, and homologous targeting ability. Cell-membrane-derived vesicles have features identical to those of their parental cells, which makes them one of the most promising materials for drug delivery. Recently, as a result of the impressive effects of immunotherapy in cancer treatment, an increasing number of researchers have used cell-membrane-derived vesicles to enhance immune responses. To be more specific, the membrane vesicles derived from immune cells, tumor cells, bacteria, or engineered cells have the antigen presentation capacity and can trigger strong anti-tumor effects of the immune system. In this review, we first indicated a brief description of the vesicles and then introduced the detection technology and drug-loading methods for them. Secondly, we concluded the characteristics and applications of vesicles derived from different sources in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuai Xiao
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chunyu Xue
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuchong Wang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
20
|
Jia X, Wang L, Feng X, Liu W, Wang X, Li F, Liu X, Yu J, Yu B, Yu X. Cell Membrane-Coated Oncolytic Adenovirus for Targeted Treatment of Glioblastoma. NANO LETTERS 2023; 23:11120-11128. [PMID: 38032110 DOI: 10.1021/acs.nanolett.3c03516] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
An oncolytic virus is a promising strategy for glioblastoma (GBM) therapy. However, there are still some challenges such as the blood-brain barrier (BBB) and preexisting immunity for targeted treatment of GBM with an oncolytic virus. In this study, two kinds of cell membrane-coated oncolytic adenoviruses (NCM-Ad and GCM-Ad) were prepared using neural stem cells (NSCs) and GBM cells as sources of membranes, respectively, and were shown to improve the targeted infectivity on GBM cells and avoid the immune clearance of preexisting neutralizing antibodies in vitro and in vivo. Specifically, NCM-Ad showed a strong ability to cross the BBB and target tumor cells in vivo. To improve the cytotoxicity to GBM, a capsid dual-modified oncolytic adenovirus (A4/k37) was also encapsulated, and NCM-A4/k37 showed outstanding tumor targeting and inhibition capacity in an orthotopic xenograft tumor model of GBM upon intravenous administration. This study provides a promising oncolytic virus-based targeted therapeutic strategy for glioma.
Collapse
Affiliation(s)
- Xinyuan Jia
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lizheng Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinyao Feng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenmo Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xupu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Fangshen Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinyao Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jiahao Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
21
|
Liu W, Zhao Y, Liu Z, Zhang G, Wu H, Zheng X, Tang X, Chen Z. Therapeutic effects against high-grade glioblastoma mediated by engineered induced neural stem cells combined with GD2-specific CAR-NK. Cell Oncol (Dordr) 2023; 46:1747-1762. [PMID: 37420122 DOI: 10.1007/s13402-023-00842-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 07/09/2023] Open
Abstract
PURPOSE High-grade glioblastoma is extremely challenging to treat because of its aggressiveness and resistance to conventional chemo- and radio-therapies. On the contrary, genetic and cellular immunotherapeutic strategies based on the stem and immune cells are emerging as promising treatments against glioblastoma (GBM). We aimed to developed a novel combined immunotherapeutic strategy to improve the treatment efficacy using genetically engineered PBMC-derived induced neural stem cells (iNSCs) expressing HSV-TK and second-generation CAR-NK cells against GBM. METHODS iNSCs cells expressing HSV-TK (iNSCsTK) and GD2-specific CAR-NK92 (GD2NK92) were generated from PBMC-derived iNSCs and NK92 cell lines, respectively. The anti-tumor effect of iNSCsTK and the combinational therapeutics of iNSCsTK and GD2NK92 were evaluated by GBM cell line using in vitro and in vivo experiments. RESULTS PBMC-derived iNSCsTK possessed tumor-tropism migration ability in vitro and in vivo, which exhibited considerable anti-tumor activity via bystander effect in the presence of ganciclovir (GCV). iNSCsTK/GCV could slow GBM progression and prolong median survival in tumor-bearing mice. However, the anti-tumor effect was limited to single therapy. Therefore, the combinational therapeutic effect of iNSCsTK/GCV and GD2NK92 against GBM was investigated. This approach displayed a more significant anti-tumor effect in vitro and in xenograft tumor mice. CONCLUSIONS PBMC-derived iNSCsTK showed a significant tumor-tropic migration and an effective anti-tumor activity with GCV in vitro and in vivo. In addition, combined with GD2NK92, iNSCsTK therapeutic efficacy improved dramatically to prolong the tumor-bearing animal model's median survival.
Collapse
Affiliation(s)
- Weihua Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Yu Zhao
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Zhongfeng Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Guangji Zhang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Huantong Wu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Xin Zheng
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Xihe Tang
- Neurosurgery Center of Aeronautical General Hospital, Beijing, 100012, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China.
- , Beijing, China.
| |
Collapse
|
22
|
Tolboom N, Verger A, Albert NL, Brendel M, Cecchin D, Fernandez PA, Fraioli F, Guedj E, Herrmann K, Traub-Weidinger T, Morbelli S, Yakushev I, Zucchetta P, Barthel H, Van Weehaeghe D. EANM position paper: theranostics in brain tumours-the present and the future. Eur J Nucl Med Mol Imaging 2023; 51:202-205. [PMID: 37698647 DOI: 10.1007/s00259-023-06425-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Affiliation(s)
- Nelleke Tolboom
- Department of Radiology and Nuclear Medicine, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, IADI, Inserm, UMR 1254, Université de Lorraine, CHRU-Nancy, Nancy, France
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine - DIMED, University Hospital of Padua, Padua, Italy
| | - Pablo Aguiar Fernandez
- Department of Radiology, Faculty of Medicine and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela, Galicia, Spain
| | - Francesco Fraioli
- Institute of Nuclear Medicine, University College London (UCL), London, UK
| | - Eric Guedj
- Département de Médecine Nucléaire, Aix Marseille Univ, APHM, CNRS, Centrale Marseille, Institut Fresnel, Hôpital de La Timone Hospital, CERIMED, Marseille, France
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)- University Hospital Essen, Essen, Germany
| | - Tatjana Traub-Weidinger
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Silvia Morbelli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Nuclear Medicine Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Igor Yakushev
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich and Munich Center for Neurosciences - Brain and Mind, Munich, Germany
| | - Pietro Zucchetta
- Department of Nuclear Medicine, University Hospital Of Padova, Padova, Italy
| | - Henryk Barthel
- Department of Nuclear Medicine, Leipzig University Medical Centre, Leipzig, Germany
| | - Donatienne Van Weehaeghe
- Department of Radiology and Nuclear Medicine, Ghent University Hospital, C. Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
23
|
Kitzberger C, Shehzad K, Morath V, Spellerberg R, Ranke J, Steiger K, Kälin RE, Multhoff G, Eiber M, Schilling F, Glass R, Weber WA, Wagner E, Nelson PJ, Spitzweg C. Interleukin-6-controlled, mesenchymal stem cell-based sodium/iodide symporter gene therapy improves survival of glioblastoma-bearing mice. Mol Ther Oncolytics 2023; 30:238-253. [PMID: 37701849 PMCID: PMC10493263 DOI: 10.1016/j.omto.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/11/2023] [Indexed: 09/14/2023] Open
Abstract
New treatment strategies are urgently needed for glioblastoma (GBM)-a tumor resistant to standard-of-care treatment with a high risk of recurrence and extremely poor prognosis. Based on their intrinsic tumor tropism, adoptively applied mesenchymal stem cells (MSCs) can be harnessed to deliver the theranostic sodium/iodide symporter (NIS) deep into the tumor microenvironment. Interleukin-6 (IL-6) is a multifunctional, highly expressed cytokine in the GBM microenvironment including recruited MSCs. MSCs engineered to drive NIS expression in response to IL-6 promoter activation offer the possibility of a new tumor-targeted gene therapy approach of GBM. Therefore, MSCs were stably transfected with an NIS-expressing plasmid controlled by the human IL-6 promoter (IL-6-NIS-MSCs) and systemically applied in mice carrying orthotopic GBM. Enhanced radiotracer uptake by 18F-Tetrafluoroborate-PET/magnetic resonance imaging (MRI) was detected in tumors after IL-6-NIS-MSC application as compared with mice that received wild-type MSCs. Ex vivo analysis of tumors and non-target organs showed tumor-specific NIS protein expression. Subsequent 131I therapy after IL-6-NIS-MSC application resulted in significantly delayed tumor growth assessed by MRI and improved median survival up to 60% of GBM-bearing mice as compared with controls. In conclusion, the application of MSC-mediated NIS gene therapy focusing on IL-6 biology-induced NIS transgene expression represents a promising approach for GBM treatment.
Collapse
Affiliation(s)
- Carolin Kitzberger
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Khuram Shehzad
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Volker Morath
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rebekka Spellerberg
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julius Ranke
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Roland E. Kälin
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Gabriele Multhoff
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Radiation Immuno-Oncology Group, Munich, Germany
- Department of Radiation Oncology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Franz Schilling
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang A. Weber
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Centre for System-Based Drug Research and Centre for Nanoscience, LMU Munich, Munich, Germany
| | - Peter J. Nelson
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christine Spitzweg
- Department of Internal Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
24
|
Nehama D, Woodell AS, Maingi SM, Hingtgen SD, Dotti G. Cell-based therapies for glioblastoma: Promising tools against tumor heterogeneity. Neuro Oncol 2023; 25:1551-1562. [PMID: 37179459 PMCID: PMC10484163 DOI: 10.1093/neuonc/noad092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Indexed: 05/15/2023] Open
Abstract
Glioblastoma (GBM) is a highly aggressive tumor with a devastating impact on quality-of-life and abysmal survivorship. Patients have very limited effective treatment options. The successes of targeted small molecule drugs and immune checkpoint inhibitors seen in various solid tumors have not translated to GBM, despite significant advances in our understanding of its molecular, immune, and microenvironment landscapes. These discoveries, however, have unveiled GBM's incredible heterogeneity and its role in treatment failure and survival. Novel cellular therapy technologies are finding successes in oncology and harbor characteristics that make them uniquely suited to overcome challenges posed by GBM, such as increased resistance to tumor heterogeneity, modularity, localized delivery, and safety. Considering these advantages, we compiled this review article on cellular therapies for GBM, focusing on cellular immunotherapies and stem cell-based therapies, to evaluate their utility. We categorize them based on their specificity, review their preclinical and clinical data, and extract valuable insights to help guide future cellular therapy development.
Collapse
Affiliation(s)
- Dean Nehama
- Department of Internal Medicine, Montefiore Medical Center, New York, New York, USA
| | - Alex S Woodell
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Spencer M Maingi
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
25
|
Li Y, Fang B. Neural stem cell-derived extracellular vesicles: The light of central nervous system diseases. Biomed Pharmacother 2023; 165:115092. [PMID: 37406512 DOI: 10.1016/j.biopha.2023.115092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023] Open
Abstract
Central nervous system (CNS) diseases are the leading cause of death worldwide. By performing compensatory functions and improving the inflammatory microenvironment, the transplantation of neural stem cells (NSCs) can promote functional recovery from brain injury, aging, brain tumours, and other diseases. However, the ability of NSCs to differentiate into neurons is limited, and they are associated with a risk of tumourigenicity. NSC-derived extracellular vesicles (NSC-EVs) can modulate the local microenvironment of the nervous system as well as distant neuronal functions. Thus, cell-free therapy may be a novel remedy for CNS disorders. This article reviews the characteristics, contents, and mechanisms of action of NSC-EVs as well as their roles and application prospects in various CNS diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
26
|
Ahmed W, Kuniyan MS, Jawed AM, Chen L. Engineered Extracellular Vesicles for Drug Delivery in Therapy of Stroke. Pharmaceutics 2023; 15:2173. [PMID: 37765144 PMCID: PMC10537154 DOI: 10.3390/pharmaceutics15092173] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
Extracellular vesicles (EVs) are promising therapeutic modalities for treating neurological conditions. EVs facilitate intercellular communication among brain cells under normal and abnormal physiological conditions. The potential capability of EVs to pass through the blood-brain barrier (BBB) makes them highly promising as nanocarrier contenders for managing stroke. EVs possess several potential advantages compared to existing drug-delivery vehicles. These advantages include their capacity to surpass natural barriers, target specific cells, and stability within the circulatory system. This review explores the trafficking and cellular uptake of EVs and evaluates recent findings in the field of EVs research. Additionally, an overview is provided of the techniques researchers utilize to bioengineer EVs for stroke therapy, new results on EV-BBB interactions, and the limitations and prospects of clinically using EVs for brain therapies. The primary objective of this study is to provide a comprehensive analysis of the advantages and challenges related to engineered EVs drug delivery, specifically focusing on their application in the treatment of stroke.
Collapse
Affiliation(s)
- Waqas Ahmed
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, China;
- School of Medicine, Southeast University, Nanjing 210009, China; (M.S.K.); (A.M.J.)
| | | | - Aqil Mohammad Jawed
- School of Medicine, Southeast University, Nanjing 210009, China; (M.S.K.); (A.M.J.)
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, China;
| |
Collapse
|
27
|
Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther 2023; 8:293. [PMID: 37544972 PMCID: PMC10404590 DOI: 10.1038/s41392-023-01536-y] [Citation(s) in RCA: 217] [Impact Index Per Article: 108.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/08/2023] Open
Abstract
Cancer remains a highly lethal disease in the world. Currently, either conventional cancer therapies or modern immunotherapies are non-tumor-targeted therapeutic approaches that cannot accurately distinguish malignant cells from healthy ones, giving rise to multiple undesired side effects. Recent advances in nanotechnology, accompanied by our growing understanding of cancer biology and nano-bio interactions, have led to the development of a series of nanocarriers, which aim to improve the therapeutic efficacy while reducing off-target toxicity of the encapsulated anticancer agents through tumor tissue-, cell-, or organelle-specific targeting. However, the vast majority of nanocarriers do not possess hierarchical targeting capability, and their therapeutic indices are often compromised by either poor tumor accumulation, inefficient cellular internalization, or inaccurate subcellular localization. This Review outlines current and prospective strategies in the design of tumor tissue-, cell-, and organelle-targeted cancer nanomedicines, and highlights the latest progress in hierarchical targeting technologies that can dynamically integrate these three different stages of static tumor targeting to maximize therapeutic outcomes. Finally, we briefly discuss the current challenges and future opportunities for the clinical translation of cancer nanomedicines.
Collapse
Affiliation(s)
- Dahua Fan
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China.
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yajun Wang
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China
| | | | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
28
|
García-Montaño LA, Licón-Muñoz Y, Martinez FJ, Keddari YR, Ziemke MK, Chohan MO, Piccirillo SG. Dissecting Intra-tumor Heterogeneity in the Glioblastoma Microenvironment Using Fluorescence-Guided Multiple Sampling. Mol Cancer Res 2023; 21:755-767. [PMID: 37255362 PMCID: PMC10390891 DOI: 10.1158/1541-7786.mcr-23-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/25/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
The treatment of the most aggressive primary brain tumor in adults, glioblastoma (GBM), is challenging due to its heterogeneous nature, invasive potential, and poor response to chemo- and radiotherapy. As a result, GBM inevitably recurs and only a few patients survive 5 years post-diagnosis. GBM is characterized by extensive phenotypic and genetic heterogeneity, creating a diversified genetic landscape and a network of biological interactions between subclones, ultimately promoting tumor growth and therapeutic resistance. This includes spatial and temporal changes in the tumor microenvironment, which influence cellular and molecular programs in GBM and therapeutic responses. However, dissecting phenotypic and genetic heterogeneity at spatial and temporal levels is extremely challenging, and the dynamics of the GBM microenvironment cannot be captured by analysis of a single tumor sample. In this review, we discuss the current research on GBM heterogeneity, in particular, the utility and potential applications of fluorescence-guided multiple sampling to dissect phenotypic and genetic intra-tumor heterogeneity in the GBM microenvironment, identify tumor and non-tumor cell interactions and novel therapeutic targets in areas that are key for tumor growth and recurrence, and improve the molecular classification of GBM.
Collapse
Affiliation(s)
- Leopoldo A. García-Montaño
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yamhilette Licón-Muñoz
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Frank J. Martinez
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| | - Yasine R. Keddari
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of California, Merced, California
| | - Michael K. Ziemke
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Muhammad O. Chohan
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sara G.M. Piccirillo
- The Brain Tumor Translational Laboratory, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
- University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| |
Collapse
|
29
|
Moleirinho S, Kitamura Y, Borges PSGN, Auduong S, Kilic S, Deng D, Kanaya N, Kozono D, Zhou J, Gray JJ, Revai-Lechtich E, Zhu Y, Shah K. Fate and Efficacy of Engineered Allogeneic Stem Cells Targeting Cell Death and Proliferation Pathways in Primary and Brain Metastatic Lung Cancer. Stem Cells Transl Med 2023; 12:444-458. [PMID: 37311043 PMCID: PMC10346421 DOI: 10.1093/stcltm/szad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/07/2023] [Indexed: 06/15/2023] Open
Abstract
Primary and metastatic lung cancer is a leading cause of cancer-related death and novel therapies are urgently needed. Epidermal growth factor receptor (EGFR) and death receptor (DR) 4/5 are both highly expressed in primary and metastatic non-small cell lung cancer (NSCLC); however, targeting these receptors individually has demonstrated limited therapeutic benefit in patients. In this study, we created and characterized diagnostic and therapeutic stem cells (SC), expressing EGFR-targeted nanobody (EV) fused to the extracellular domain of death DR4/5 ligand (DRL) (EVDRL) that simultaneously targets EGFR and DR4/5, in primary and metastatic NSCLC tumor models. We show that EVDRL targets both cell surface receptors, and induces caspase-mediated apoptosis in a broad spectrum of NSCLC cell lines. Utilizing real-time dual imaging and correlative immunohistochemistry, we show that allogeneic SCs home to tumors and when engineered to express EVDRL, alleviate tumor burden and significantly increase survival in primary and brain metastatic NSCLC. This study reports mechanistic insights into simultaneous targeting of EGFR- and DR4/5 in lung tumors and presents a promising approach for translation into the clinical setting.
Collapse
Affiliation(s)
- Susana Moleirinho
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Yohei Kitamura
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Paulo S G N Borges
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sophia Auduong
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Seyda Kilic
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - David Deng
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nobuhiko Kanaya
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - David Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jing Zhou
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MA, USA
| | - Jeffrey J Gray
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MA, USA
| | - Esther Revai-Lechtich
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Yanni Zhu
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
30
|
Russo MN, Whaley LA, Norton ES, Zarco N, Guerrero-Cázares H. Extracellular vesicles in the glioblastoma microenvironment: A diagnostic and therapeutic perspective. Mol Aspects Med 2023; 91:101167. [PMID: 36577547 PMCID: PMC10073317 DOI: 10.1016/j.mam.2022.101167] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM), is the most malignant form of gliomas and the most common and lethal primary brain tumor in adults. Conventional cancer treatments have limited to no efficacy on GBM. GBM cells respond and adapt to the surrounding brain parenchyma known as tumor microenvironment (TME) to promote tumor preservation. Among specific TME, there are 3 of particular interest for GBM biology: the perivascular niche, the subventricular zone neurogenic niche, and the immune microenvironment. GBM cells and TME cells present a reciprocal feedback which results in tumor maintenance. One way that these cells can communicate is through extracellular vesicles. These vesicles include exosomes and microvesicles that have the ability to carry both cancerous and non-cancerous cargo, such as miRNA, RNA, proteins, lipids, and DNA. In this review we will discuss the booming topic that is extracellular vesicles, and how they have the novelty to be a diagnostic and targetable vehicle for GBM.
Collapse
Affiliation(s)
- Marissa N Russo
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Lauren A Whaley
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA; Biology Graduate Program, University of North Florida, Jacksonville, FL, USA
| | - Emily S Norton
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA; Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Natanael Zarco
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
31
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
32
|
Morgenroth A, Baazaoui F, Hosseinnejad A, Schäfer L, Vogg A, Singh S, Mottaghy FM. Neural Stem Cells as Carriers of Nucleoside-Conjugated Nanogels: A New Approach toward Cell-Mediated Delivery. ACS APPLIED MATERIALS & INTERFACES 2023; 15:21792-21803. [PMID: 37127284 PMCID: PMC10176478 DOI: 10.1021/acsami.2c23283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Neural stem cells (NSCs) present attractive natural drug delivery systems (DDSs). Their migratory potential enables crossing of the blood-brain barrier and efficient and selective accumulation near malignant cells. Here, we present the potential of NSCs as DDSs for nucleoside analogue-conjugated nanogels (NGs). Two different approaches were investigated: the intracellular loading and extracellular cell surface decoration with NGs. For both designs, the tumor-specific migratory potentials of NSCs remained unchanged; however, the intracellular loading showed a shorter NG retention. The cell surface decoration protocol yielded a high loading capacity of 100% after 1 h and a prolonged drug retention. A redox-sensitive linker between NGs and the nucleoside analogue 5-ethynyl-2'-deoxycytidine (EdC) allowed a tumor environment-specific drug release and its efficient and preferential incorporation into the DNA of the tumor cells. Interestingly, the tumor-trafficking potentials of NSCs were significantly potentiated by irradiation of tumor cells. In conclusion, this study indicates the potentials of cell surface-decorated NSCs as DDSs for tumor-specific release, cellular uptake, and incorporation of EdC into DNA.
Collapse
Affiliation(s)
| | - Fatima Baazaoui
- Department of Nuclear Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Aisa Hosseinnejad
- DWI - Leibniz-Institute for Interactive Materials, RWTH Aachen University, 52074 Aachen, Germany
| | - Laura Schäfer
- Department of Nuclear Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Andreas Vogg
- Department of Nuclear Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | - Smriti Singh
- DWI - Leibniz-Institute for Interactive Materials, RWTH Aachen University, 52074 Aachen, Germany
- Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH Aachen University, 52074 Aachen, Germany
- Department of Nuclear Medicine, Maastricht University Medical Centre, 6229 HX Maastricht, Netherlands
| |
Collapse
|
33
|
Ou A, Wang Y, Zhang J, Huang Y. Living Cells and Cell-Derived Vesicles: A Trojan Horse Technique for Brain Delivery. Pharmaceutics 2023; 15:pharmaceutics15041257. [PMID: 37111742 PMCID: PMC10145830 DOI: 10.3390/pharmaceutics15041257] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Brain diseases remain a significant global healthcare burden. Conventional pharmacological therapy for brain diseases encounters huge challenges because of the blood-brain barrier (BBB) limiting the delivery of therapeutics into the brain parenchyma. To address this issue, researchers have explored various types of drug delivery systems. Cells and cell derivatives have attracted increasing interest as "Trojan horse" delivery systems for brain diseases, owing to their superior biocompatibility, low immunogenicity, and BBB penetration properties. This review provided an overview of recent advancements in cell- and cell-derivative-based delivery systems for the diagnosis and treatment of brain diseases. Additionally, it discussed the challenges and potential solutions for clinical translation.
Collapse
Affiliation(s)
- Ante Ou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuewei Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
34
|
Loras A, Gonzalez-Bonet LG, Gutierrez-Arroyo JL, Martinez-Cadenas C, Marques-Torrejon MA. Neural Stem Cells as Potential Glioblastoma Cells of Origin. Life (Basel) 2023; 13:life13040905. [PMID: 37109434 PMCID: PMC10145968 DOI: 10.3390/life13040905] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor in adults and it remains incurable. These tumors are very heterogeneous, resistant to cytotoxic therapies, and they show high rates of invasiveness. Therefore, patients face poor prognosis, and the survival rates remain very low. Previous research states that GBM contains a cell population with stem cell characteristics called glioma stem cells (GSCs). These cells are able to self-renew and regenerate the tumor and, therefore, they are partly responsible for the observed resistance to therapies and tumor recurrence. Recent data indicate that neural stem cells (NSCs) in the subventricular zone (SVZ) are the cells of origin of GBM, that is, the cell type acquiring the initial tumorigenic mutation. The involvement of SVZ-NSCs is also associated with GBM progression and recurrence. Identifying the cellular origin of GBM is important for the development of early detection techniques and the discovery of early disease markers. In this review, we analyze the SVZ-NSC population as a potential GBM cell of origin, and its potential role for GBM therapies.
Collapse
Affiliation(s)
- Alba Loras
- Department of Medicine, University of Valencia, 46010 Valencia, Spain
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon de la Plana, Spain
| | - Julia L. Gutierrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon de la Plana, Spain
| | | | - Maria Angeles Marques-Torrejon
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon de la Plana, Spain
- Correspondence: ; Tel.: +34-964-387-478
| |
Collapse
|
35
|
Ghasemi Darestani N, Gilmanova AI, Al-Gazally ME, Zekiy AO, Ansari MJ, Zabibah RS, Jawad MA, Al-Shalah SAJ, Rizaev JA, Alnassar YS, Mohammed NM, Mustafa YF, Darvishi M, Akhavan-Sigari R. Mesenchymal stem cell-released oncolytic virus: an innovative strategy for cancer treatment. Cell Commun Signal 2023; 21:43. [PMID: 36829187 PMCID: PMC9960453 DOI: 10.1186/s12964-022-01012-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/10/2022] [Indexed: 02/26/2023] Open
Abstract
Oncolytic viruses (OVs) infect, multiply, and finally remove tumor cells selectively, causing no damage to normal cells in the process. Because of their specific features, such as, the ability to induce immunogenic cell death and to contain curative transgenes in their genomes, OVs have attracted attention as candidates to be utilized in cooperation with immunotherapies for cancer treatment. This treatment takes advantage of most tumor cells' inherent tendency to be infected by certain OVs and both innate and adaptive immune responses are elicited by OV infection and oncolysis. OVs can also modulate tumor microenvironment and boost anti-tumor immune responses. Mesenchymal stem cells (MSC) are gathering interest as promising anti-cancer treatments with the ability to address a wide range of cancers. MSCs exhibit tumor-trophic migration characteristics, allowing them to be used as delivery vehicles for successful, targeted treatment of isolated tumors and metastatic malignancies. Preclinical and clinical research were reviewed in this study to discuss using MSC-released OVs as a novel method for the treatment of cancer. Video Abstract.
Collapse
Affiliation(s)
| | - Anna I Gilmanova
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | | | - Angelina O Zekiy
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | | - Saif A J Al-Shalah
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Mohammad Darvishi
- Department of Aerospace and Subaquatic Medicine, Infectious Diseases and Tropical Medicine Research Center (IDTMRC), AJA University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany.,Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
36
|
Varela ML, Comba A, Faisal SM, Argento A, Franson A, Barissi MN, Sachdev S, Castro MG, Lowenstein PR. Gene Therapy for High Grade Glioma: The Clinical Experience. Expert Opin Biol Ther 2023; 23:145-161. [PMID: 36510843 PMCID: PMC9998375 DOI: 10.1080/14712598.2022.2157718] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION High-grade gliomas (HGG) are the most common malignant primary brain tumors in adults, with a median survival of ~18 months. The standard of care (SOC) is maximal safe surgical resection, and radiation therapy with concurrent and adjuvant temozolomide. This protocol remains unchanged since 2005, even though HGG median survival has marginally improved. AREAS COVERED Gene therapy was developed as a promising approach to treat HGG. Here, we review completed and ongoing clinical trials employing viral and non-viral vectors for adult and pediatric HGG, as well as the key supporting preclinical data. EXPERT OPINION These therapies have proven safe, and pre- and post-treatment tissue analyses demonstrated tumor cell lysis, increased immune cell infiltration, and increased systemic immune function. Although viral therapy in clinical trials has not yet significantly extended the survival of HGG, promising strategies are being tested. Oncolytic HSV vectors have shown promising results for both adult and pediatric HGG. A recently published study demonstrated that HG47Δ improved survival in recurrent HGG. Likewise, PVSRIPO has shown survival improvement compared to historical controls. It is likely that further analysis of these trials will stimulate the development of new administration protocols, and new therapeutic combinations that will improve HGG prognosis.
Collapse
Affiliation(s)
- Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anna Argento
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrea Franson
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Marcus N Barissi
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Sean Sachdev
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
37
|
Choi Y, Lee HK, Choi KC. Engineered adult stem cells: a promising tool for anti-cancer therapy. BMB Rep 2023; 56:71-77. [PMID: 36330711 PMCID: PMC9978368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/28/2022] [Accepted: 11/04/2022] [Indexed: 02/24/2023] Open
Abstract
Cancers are one of the most dreaded diseases in human history and have been targeted by numerous trials including surgery, chemotherapy, radiation therapy, and anti-cancer drugs. Adult stem cells (ASCs), which can regenerate tissues and repair damage, have emerged as leading therapeutic candidates due to their homing ability toward tumor foci. Stem cells can precisely target malicious tumors, thereby minimizing the toxicity of normal cells and unfavorable side effects. ASCs, such as mesenchymal stem cells (MSCs), neural stem cells (NSCs), and hematopoietic stem cells (HSCs), are powerful tools for delivering therapeutic agents to various primary and metastatic cancers. Engineered ASCs act as a bridge between the tumor sites and tumoricidal reagents, producing therapeutic substances such as exosomes, viruses, and anti-cancer proteins encoded by several suicide genes. This review focuses on various anti-cancer therapies implemented via ASCs and summarizes the recent treatment progress and shortcomings. [BMB Reports 2023; 56(2): 71-77].
Collapse
Affiliation(s)
- Youngdong Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Hong Kyu Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
38
|
Ma R, Li Z, Chiocca EA, Caligiuri MA, Yu J. The emerging field of oncolytic virus-based cancer immunotherapy. Trends Cancer 2023; 9:122-139. [PMID: 36402738 PMCID: PMC9877109 DOI: 10.1016/j.trecan.2022.10.003] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/18/2022]
Abstract
Oncolytic viruses (OVs) provide novel and promising therapeutic options for patients with cancers resistant to traditional therapies. Natural or genetically modified OVs are multifaceted tumor killers. They directly lyse tumor cells while sparing normal cells, and indirectly potentiate antitumor immunity by releasing antigens and activating inflammatory responses in the tumor microenvironment. However, some limitations, such as limited penetration of OVs into tumors, short persistence, and the host antiviral immune response, are impeding the broad translation of oncolytic virotherapy into the clinic. If these challenges can be overcome, combination therapies, such as OVs plus immune checkpoint blockade (ICB), chimeric antigen receptor (CAR) T cells, or CAR natural killer (NK) cells, may provide powerful therapeutic platforms in the clinic.
Collapse
Affiliation(s)
- Rui Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhenlong Li
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - E Antonio Chiocca
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA; Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA; Department of Immuno-Oncology, Beckman Research Institute, Los Angeles, CA 91010, USA.
| |
Collapse
|
39
|
Choi Y, Lee HK, Choi KC. Engineered adult stem cells: a promising tool for anti-cancer therapy. BMB Rep 2023; 56:71-77. [PMID: 36330711 PMCID: PMC9978368 DOI: 10.5483/bmbrep.2022-0091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/28/2022] [Accepted: 11/04/2022] [Indexed: 08/03/2023] Open
Abstract
Cancers are one of the most dreaded diseases in human history and have been targeted by numerous trials including surgery, chemotherapy, radiation therapy, and anti-cancer drugs. Adult stem cells (ASCs), which can regenerate tissues and repair damage, have emerged as leading therapeutic candidates due to their homing ability toward tumor foci. Stem cells can precisely target malicious tumors, thereby minimizing the toxicity of normal cells and unfavorable side effects. ASCs, such as mesenchymal stem cells (MSCs), neural stem cells (NSCs), and hematopoietic stem cells (HSCs), are powerful tools for delivering therapeutic agents to various primary and metastatic cancers. Engineered ASCs act as a bridge between the tumor sites and tumoricidal reagents, producing therapeutic substances such as exosomes, viruses, and anti-cancer proteins encoded by several suicide genes. This review focuses on various anti-cancer therapies implemented via ASCs and summarizes the recent treatment progress and shortcomings. [BMB Reports 2023; 56(2): 71-77].
Collapse
Affiliation(s)
- Youngdong Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Hong Kyu Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
40
|
Potent bystander effect and tumor tropism in suicide gene therapy using stem cells from human exfoliated deciduous teeth. Cancer Gene Ther 2023; 30:85-95. [PMID: 36076062 DOI: 10.1038/s41417-022-00527-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 08/01/2022] [Accepted: 08/24/2022] [Indexed: 01/20/2023]
Abstract
Herpes simplex virus thymidine kinase (HSVTK)/ganciclovir (GCV) suicide gene therapy has a long history of treating malignant gliomas. Recently, stem cells from human exfoliated deciduous teeth (SHED), which are collected from deciduous teeth and have excellent harvestability, ethical aspects, and self-renewal, have been attracting attention mainly in the field of gene therapy. In the present study, we assessed SHED as a novel cellular vehicle for suicide gene therapy in malignant gliomas, as we have previously demonstrated with various cell types. SHED was transduced with the HSVTK gene (SHEDTK). In vitro experiments showed a significant bystander effect between SHEDTK and glioma cell lines in coculture. Furthermore, apoptotic changes caused by caspase 3/7 activation were simultaneously observed in SHEDTK and glioma cells. Mice implanted with a mixture of U87 and SHEDTK and treated with intraperitoneal GCV survived for longer than 100 days. Additionally, tumors in treatment model mice were significantly reduced in size during the treatment period. SHEDTK implanted at the contralateral hemisphere migrated toward the tumor crossing the corpus callosum. These results suggested that SHEDTK-based suicide gene therapy has potent tumor tropism and a bystander-killing effect, potentially offering a new promising therapeutic modality for malignant gliomas.
Collapse
|
41
|
Noureldine MHA, Shimony N, Jallo GI. Malignant Spinal Tumors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1405:565-581. [PMID: 37452954 DOI: 10.1007/978-3-031-23705-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Malignant spinal tumors constitute around 22% of all primary spinal tumors. The most common location of metastases to the spinal region is the extradural compartment. The molecular and genetic characterization of these tumors was the basis for the updated WHO classification of CNS tumors in 2016, where many CNS tumors are now diagnosed according to their genetic profile rather than relying solely on the histopathological appearance. Magnetic resonance imaging (MRI) is the current gold standard for the initial evaluation and subsequent follow-up on intradural spinal cord tumors, and the imaging sequences must include T2-weighted images (WI), short time inversion recovery (STIR), and pre- and post-contrast T1-WI in the axial, sagittal, and coronal planes. The clinical presentation is highly variable and depends on the tumor size, growth rate, type, infiltrative, necrotic and hemorrhagic potential as well as the exact location within the spinal compartment. Surgical intervention remains the mainstay of management of symptomatic and radiographically enlarging spinal tumors, where the goal is to achieve maximal safe resection. Tumor recurrences are managed with repeat surgical resection (preferred whenever possible and safe), radiotherapy, chemotherapy, or any combination of these therapies.
Collapse
Affiliation(s)
- Mohammad Hassan A Noureldine
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Johns Hopkins University School of Medicine, Institute for Brain Protection Sciences, Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA
| | - Nir Shimony
- Johns Hopkins University School of Medicine, Institute for Brain Protection Sciences, Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA
- Geisinger Medical Center, Institute of Neuroscience, Geisinger Commonwealth School of Medicine, Danville, PA, USA
| | - George I Jallo
- Institute for Brain Protections Sciences, Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA.
| |
Collapse
|
42
|
Andersen BM, Reardon DA. Immunotherapy approaches for adult glioma: knowledge gained from recent clinical trials. Curr Opin Neurol 2022; 35:803-813. [PMID: 36367046 DOI: 10.1097/wco.0000000000001118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE OF REVIEW Summarize principles behind various immunotherapy approaches for high and low-grade glioma in the context of recently completed clinical trials and the new insights they provide. RECENT FINDINGS Despite the widespread success of therapies targeting the T-cell checkpoints programmed-death 1 and cytotoxic T lymphocyte antigen 4 in other malignancies, recent phase III trials in glioblastoma confirm the lack of efficacy of anti-programmed-death 1 monotherapy in more than 90% of patients. Vaccination approaches remain under investigation for high-grade glioma and have shown activity in some low-grade glioma patients. Chimeric antigen receptor T cells now feature a new generation of products engineered to potentially withstand glucocorticoid therapy. Oncolytic viral therapies have similarly advanced in sophistication, with drug-sensitive gene expression and tumor-selective modifications. Combinations of therapies hold promise for overcoming the numerous mechanisms of immune suppression in glioma. SUMMARY Although immunotherapies have yet to show rates of efficacy compared with other malignancies, new knowledge of immunology and combination therapies brings hope for improved efficacy in the future.
Collapse
Affiliation(s)
- Brian M Andersen
- Department of Neurology, Brigham and Women's Hospital
- Department of Neurology, Veterans Affairs Medical Center
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Rouzbahani E, Majidpoor J, Najafi S, Mortezaee K. Cancer stem cells in immunoregulation and bypassing anti-checkpoint therapy. Biomed Pharmacother 2022; 156:113906. [DOI: 10.1016/j.biopha.2022.113906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/26/2022] Open
|
44
|
Sim TM. Nanoparticle-assisted targeting of the tumour microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
Ahmad F, Varghese R, Panda S, Ramamoorthy S, Areeshi MY, Fagoonee S, Haque S. Smart Nanoformulations for Brain Cancer Theranostics: Challenges and Promises. Cancers (Basel) 2022; 14:5389. [PMID: 36358807 PMCID: PMC9655255 DOI: 10.3390/cancers14215389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
Despite their low prevalence, brain tumors are among the most lethal cancers. They are extremely difficult to diagnose, monitor and treat. Conventional anti-cancer strategies such as radio- and chemotherapy have largely failed, and to date, the development of even a single effective therapeutic strategy against central nervous system (CNS) tumors has remained elusive. There are several factors responsible for this. Brain cancers are a heterogeneous group of diseases with variable origins, biochemical properties and degrees of invasiveness. High-grade gliomas are amongst the most metastatic and invasive cancers, which is another reason for therapeutic failure in their case. Moreover, crossing the blood brain and the blood brain tumor barriers has been a significant hindrance in the development of efficient CNS therapeutics. Cancer nanomedicine, which encompasses the application of nanotechnology for diagnosis, monitoring and therapy of cancers, is a rapidly evolving field of translational medicine. Nanoformulations, because of their extreme versatility and manipulative potential, are emerging candidates for tumor targeting, penetration and treatment in the brain. Moreover, suitable nanocarriers can be commissioned for theranostics, a combinatorial personalized approach for simultaneous imaging and therapy. This review first details the recent advances in novel bioengineering techniques that provide promising avenues for circumventing the hurdles of delivering the diagnostic/therapeutic agent to the CNS. The authors then describe in detail the tremendous potential of utilizing nanotechnology, particularly nano-theranostics for brain cancer imaging and therapy, and outline the different categories of recently developed next-generation smart nanoformulations that have exceptional potential for making a breakthrough in clinical neuro-oncology therapeutics.
Collapse
Affiliation(s)
- Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore 632014, India
| | - Ressin Varghese
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore 632014, India
| | - Subhrajita Panda
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore 632014, India
| | - Siva Ramamoorthy
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore 632014, India
| | - Mohammad Y. Areeshi
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, 10126 Turin, Italy
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
46
|
Wang L, Shi Y, Jiang J, Li C, Zhang H, Zhang X, Jiang T, Wang L, Wang Y, Feng L. Micro-Nanocarriers Based Drug Delivery Technology for Blood-Brain Barrier Crossing and Brain Tumor Targeting Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203678. [PMID: 36103614 DOI: 10.1002/smll.202203678] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/04/2022] [Indexed: 06/15/2023]
Abstract
The greatest obstacle to using drugs to treat brain tumors is the blood-brain barrier (BBB), making it difficult for conventional drug molecules to enter the brain. Therefore, how to safely and effectively penetrate the BBB to achieve targeted drug delivery to brain tumors has been a challenging research problem. With the intensive research in micro- and nanotechnology in recent years, nano drug-targeted delivery technologies have shown great potential to overcome this challenge, such as inorganic nanocarriers, organic polymer-carriers, liposomes, and biobased carriers, which can be designed in different sizes, shapes, and surface functional groups to enhance their ability to penetrate the BBB and targeted drug delivery for brain tumors. In this review, the composition and overcoming patterns of the BBB are detailed, and then the hot research topics of drug delivery carriers for brain tumors in recent years are summarized, and their mechanisms of action on the BBB and the factors affecting drug delivery are described in detail, and the effectiveness of targeted therapy for brain tumors is evaluated. Finally, the challenges and dilemmas in developing brain tumor drug delivery systems are discussed, which will be promising in the future for targeted drug delivery to brain tumors based on micro-nanocarriers technology.
Collapse
Affiliation(s)
- Luyao Wang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Youyuan Shi
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Jingzhen Jiang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Chan Li
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Hengrui Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Xinhui Zhang
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
| | - Tao Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yinyan Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Lin Feng
- School of Mechanical Engineering & Automation, Beihang University, Beijing, 100191, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
47
|
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
|
48
|
Satterlee AB, Dunn DE, Valdivia A, Malawsky D, Buckley A, Gershon T, Floyd S, Hingtgen S. Spatiotemporal analysis of induced neural stem cell therapy to overcome advanced glioblastoma recurrence. Mol Ther Oncolytics 2022; 26:49-62. [PMID: 35784402 PMCID: PMC9217992 DOI: 10.1016/j.omto.2022.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/01/2022] [Indexed: 12/03/2022] Open
Abstract
Genetically engineered neural stem cells (NSCs) are a promising therapy for the highly aggressive brain cancer glioblastoma (GBM); however, treatment durability remains a major challenge. We sought to define the events that contribute to dynamic adaptation of GBM during treatment with human skin-derived induced NSCs releasing the pro-apoptotic agent TRAIL (iNSC-TRAIL) and develop strategies that convert initial tumor kill into sustained GBM suppression. In vivo and ex vivo analysis before, during, and after treatment revealed significant shifts in tumor transcriptome and spatial distribution as the tumors adapted to treatment. To address this, we designed iNSC delivery strategies that increased spatiotemporal TRAIL coverage and significantly decreased GBM volume throughout the brain, reducing tumor burden 100-fold as quantified in live ex vivo brain slices. The varying impact of different strategies on treatment durability and median survival of both solid and invasive tumors provides important guidance for optimizing iNSC therapy.
Collapse
Affiliation(s)
- Andrew B. Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Denise E. Dunn
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27704, 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
| | - Daniel Malawsky
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Andrew Buckley
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Timothy Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Scott Floyd
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27704, USA
| | - Shawn Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
49
|
Amadasu E, Panther E, Lucke-Wold B. Characterization and Treatment of Spinal Tumors. INTENSIVE CARE RESEARCH 2022; 2:76-95. [PMID: 36741203 PMCID: PMC9893847 DOI: 10.1007/s44231-022-00014-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/03/2022] [Indexed: 02/07/2023]
Abstract
AbstractThe prevalence of spinal tumors is rare in comparison to brain tumors which encompass most central nervous system tumors. Tumors of the spine can be divided into primary and metastatic tumors with the latter being the most common presentation. Primary tumors are subdivided based on their location on the spinal column and in the spinal cord into intramedullary, intradural extramedullary, and primary bone tumors. Back pain is a common presentation in spine cancer patients; however, other radicular pain may be present. Magnetic resonance imaging (MRI) is the imaging modality of choice for intradural extramedullary and intramedullary tumors. Plain radiographs are used in the initial diagnosis of primary bone tumors while Computed tomography (CT) and MRI may often be necessary for further characterization. Complete surgical resection is the treatment of choice for spinal tumors and may be curative for well circumscribed lesions. However, intralesional resection along with adjuvant radiation and chemotherapy can be indicated for patients that would experience increased morbidity from damage to nearby neurological structures caused by resection with wide margins. Even with the current treatment options, the prognosis for aggressive spinal cancer remains poor. Advances in novel treatments including molecular targeting, immunotherapy and stem cell therapy provide the potential for greater control of malignant and metastatic tumors of the spine.
Collapse
|
50
|
López-Ornelas A, Jiménez A, Pérez-Sánchez G, Rodríguez-Pérez CE, Corzo-Cruz A, Velasco I, Estudillo E. The Impairment of Blood-Brain Barrier in Alzheimer's Disease: Challenges and Opportunities with Stem Cells. Int J Mol Sci 2022; 23:ijms231710136. [PMID: 36077533 PMCID: PMC9456198 DOI: 10.3390/ijms231710136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/28/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder and its prevalence is increasing. Nowadays, very few drugs effectively reduce AD symptoms and thus, a better understanding of its pathophysiology is vital to design new effective schemes. Presymptomatic neuronal damage caused by the accumulation of Amyloid β peptide and Tau protein abnormalities remains a challenge, despite recent efforts in drug development. Importantly, therapeutic targets, biomarkers, and diagnostic techniques have emerged to detect and treat AD. Of note, the compromised blood-brain barrier (BBB) and peripheral inflammation in AD are becoming more evident, being harmful factors that contribute to the development of the disease. Perspectives from different pre-clinical and clinical studies link peripheral inflammation with the onset and progression of AD. This review aims to analyze the main factors and the contribution of impaired BBB in AD development. Additionally, we describe the potential therapeutic strategies using stem cells for AD treatment.
Collapse
Affiliation(s)
- Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico
- Hospital Nacional Homeopático, Hospitales Federales de Referencia, Mexico City 06800, Mexico
| | - Adriana Jiménez
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Calzada México-Xochimilco 101, Colonia San Lorenzo Huipulco, Tlalpan, Ciudad de México 14370, Mexico
| | - Citlali Ekaterina Rodríguez-Pérez
- Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico
| | - Alejandro Corzo-Cruz
- Laboratorio Traslacional, Escuela Militar de Graduados de Sanidad, Secretaría de la Defensa Nacional, Batalla de Celaya 202, Lomas de Sotelo, Miguel Hidalgo, Ciudad de México 11200, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular—Neurociencias, Universidad Nacional Autónoma de Mexico, Mexico City 04510, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico
- Correspondence:
| |
Collapse
|