51
|
Gečys D, Skredėnienė R, Gečytė E, Kazlauskas A, Balnytė I, Jekabsone A. Adipose Tissue-Derived Stem Cell Extracellular Vesicles Suppress Glioblastoma Proliferation, Invasiveness and Angiogenesis. Cells 2023; 12:cells12091247. [PMID: 37174646 PMCID: PMC10177295 DOI: 10.3390/cells12091247] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Extracellular vesicles (EVs) are attractive anticancer drug delivery candidates as they confer several fundamental properties, such as low immunogenicity and the ability to cross biological barriers. Mesenchymal stem cells (MSCs) are convenient producers for high EV yields, and patient-derived adipose tissue MSC-EVs could serve as personalised carriers. However, MSC-EV applications raise critical concerns as their natural cargo can affect tumour progression in both inducing and suppressing ways. In this study, we investigated the effect of adipose tissue-derived mesenchymal stem cell EVs (ASC-EVs) on several glioblastoma (GBM) cell lines to define their applicability for anticancer therapies. ASC-EVs were isolated from a cell-conditioned medium and characterised by size and specific markers. The internalisation of fluorescently labelled ASC-EVs by human GBM cells HROG36, U87 MG, and T98G was evaluated by fluorescent microscopy. Changes in GBM cell proliferation after ASC-EV application were determined by the metabolic PrestoBlue assay. Expression alterations in genes responsible for cell adhesion, proliferation, migration, and angiogenesis were evaluated by quantitative real-time PCR. ASC-EV effects on tumour invasiveness and neoangiogenesis in ovo were analysed on the chicken embryo chorioallantoic membrane model (CAM). ASC-EV treatment reduced GBM proliferation in vitro and significantly downregulated invasiveness-related genes ITGα5 (in T98G and HROG63) and ITGβ3 (in HROG36) and the vascularisation-inducing gene KDR (in all GBM lines). Additionally, an approximate 65% reduction in the GBM invasion rate was observed in CAM after ASC-EV treatment. Our study indicates that ASC-EVs possess antitumour properties, reducing GBM cell proliferation and invasiveness, and can be applied as anticancer therapeutics and medicine carriers.
Collapse
Affiliation(s)
- Dovydas Gečys
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
| | - Rūta Skredėnienė
- Department of Histology and Embryology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Emilija Gečytė
- Laboratory of Molecular Cardiology, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
| | - Arūnas Kazlauskas
- Laboratory of Molecular Neurooncology, Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania
| | - Ingrida Balnytė
- Department of Histology and Embryology, Faculty of Medicine, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Aistė Jekabsone
- Institute of Pharmaceutical Technologies, Faculty of Pharmacy, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
- Preclinical Research Laboratory for Medicinal Products, Institute of Cardiology, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
| |
Collapse
|
52
|
Wang C, Li Y, Gu L, Chen R, Zhu H, Zhang X, Zhang Y, Feng S, Qiu S, Jian Z, Xiong X. Gene Targets of CAR-T Cell Therapy for Glioblastoma. Cancers (Basel) 2023; 15:cancers15082351. [PMID: 37190280 DOI: 10.3390/cancers15082351] [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: 02/27/2023] [Revised: 04/05/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with a poor prognosis following conventional therapeutic interventions. Moreover, the blood-brain barrier (BBB) severely impedes the permeation of chemotherapy drugs, thereby reducing their efficacy. Consequently, it is essential to develop novel GBM treatment methods. A novel kind of pericyte immunotherapy known as chimeric antigen receptor T (CAR-T) cell treatment uses CAR-T cells to target and destroy tumor cells without the aid of the antigen with great specificity and in a manner that is not major histocompatibility complex (MHC)-restricted. It has emerged as one of the most promising therapy techniques with positive clinical outcomes in hematological cancers, particularly leukemia. Due to its efficacy in hematologic cancers, CAR-T cell therapy could potentially treat solid tumors, including GBM. On the other hand, CAR-T cell treatment has not been as therapeutically effective in treating GBM as it has in treating other hematologic malignancies. CAR-T cell treatments for GBM have several challenges. This paper reviewed the use of CAR-T cell therapy in hematologic tumors and the selection of targets, difficulties, and challenges in GBM.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
| | - Yuntao Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ran Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xu Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shi Feng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Qiu
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou 313003, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
| |
Collapse
|
53
|
Said SS, Ibrahim WN. Cancer Resistance to Immunotherapy: Comprehensive Insights with Future Perspectives. Pharmaceutics 2023; 15:pharmaceutics15041143. [PMID: 37111629 PMCID: PMC10141036 DOI: 10.3390/pharmaceutics15041143] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Cancer immunotherapy is a type of treatment that harnesses the power of the immune systems of patients to target cancer cells with better precision compared to traditional chemotherapy. Several lines of treatment have been approved by the US Food and Drug Administration (FDA) and have led to remarkable success in the treatment of solid tumors, such as melanoma and small-cell lung cancer. These immunotherapies include checkpoint inhibitors, cytokines, and vaccines, while the chimeric antigen receptor (CAR) T-cell treatment has shown better responses in hematological malignancies. Despite these breakthrough achievements, the response to treatment has been variable among patients, and only a small percentage of cancer patients gained from this treatment, depending on the histological type of tumor and other host factors. Cancer cells develop mechanisms to avoid interacting with immune cells in these circumstances, which has an adverse effect on how effectively they react to therapy. These mechanisms arise either due to intrinsic factors within cancer cells or due other cells within the tumor microenvironment (TME). When this scenario is used in a therapeutic setting, the term “resistance to immunotherapy” is applied; “primary resistance” denotes a failure to respond to treatment from the start, and “secondary resistance” denotes a relapse following the initial response to immunotherapy. Here, we provide a thorough summary of the internal and external mechanisms underlying tumor resistance to immunotherapy. Furthermore, a variety of immunotherapies are briefly discussed, along with recent developments that have been employed to prevent relapses following treatment, with a focus on upcoming initiatives to improve the efficacy of immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Sawsan Sudqi Said
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
54
|
Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer 2023; 9:270-292. [PMID: 36681605 PMCID: PMC10038906 DOI: 10.1016/j.trecan.2022.12.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in understanding tumor biology, malignant gliomas remain incurable. While immunotherapy has improved outcomes in other cancer types, comparable efficacy has not yet been demonstrated for primary cancers of the central nervous system (CNS). T cell exhaustion, defined as a progressive decrease in effector function, sustained expression of inhibitory receptors, metabolic dysfunction, and distinct epigenetic and transcriptional alterations, contributes to the failure of immunotherapy in the CNS. Herein, we describe recent advances in understanding the drivers of T cell exhaustion in the glioma microenvironment. We discuss the extrinsic and intrinsic factors that contribute to exhaustion and highlight potential avenues for reversing this phenotype. Our ability to directly target specific immunosuppressive drivers in brain cancers would be a major advance in immunotherapy.
Collapse
Affiliation(s)
- Matthew B Watowich
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
55
|
Mercogliano MF, Bruni S, Mauro FL, Schillaci R. Emerging Targeted Therapies for HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15071987. [PMID: 37046648 PMCID: PMC10093019 DOI: 10.3390/cancers15071987] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and the leading cause of death. HER2 overexpression is found in approximately 20% of breast cancers and is associated with a poor prognosis and a shorter overall survival. Tratuzumab, a monoclonal antibody directed against the HER2 receptor, is the standard of care treatment. However, a third of the patients do not respond to therapy. Given the high rate of resistance, other HER2-targeted strategies have been developed, including monoclonal antibodies such as pertuzumab and margetuximab, trastuzumab-based antibody drug conjugates such as trastuzumab-emtansine (T-DM1) and trastuzumab-deruxtecan (T-DXd), and tyrosine kinase inhibitors like lapatinib and tucatinib, among others. Moreover, T-DXd has proven to be of use in the HER2-low subtype, which suggests that other HER2-targeted therapies could be successful in this recently defined new breast cancer subclassification. When patients progress to multiple strategies, there are several HER2-targeted therapies available; however, treatment options are limited, and the potential combination with other drugs, immune checkpoint inhibitors, CAR-T cells, CAR-NK, CAR-M, and vaccines is an interesting and appealing field that is still in development. In this review, we will discuss the highlights and pitfalls of the different HER2-targeted therapies and potential combinations to overcome metastatic disease and resistance to therapy.
Collapse
|
56
|
Guzman G, Pellot K, Reed MR, Rodriguez A. CAR T-cells to treat brain tumors. Brain Res Bull 2023; 196:76-98. [PMID: 36841424 DOI: 10.1016/j.brainresbull.2023.02.014] [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: 07/11/2022] [Revised: 01/18/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
Tremendous success using CAR T therapy in hematological malignancies has garnered significant interest in developing such treatments for solid tumors, including brain tumors. This success, however, has yet to be mirrored in solid organ neoplasms. CAR T function has shown limited efficacy against brain tumors due to several factors including the immunosuppressive tumor microenvironment, blood-brain barrier, and tumor-antigen heterogeneity. Despite these considerations, CAR T-cell therapy has the potential to be implemented as a treatment modality for brain tumors. Here, we review adult and pediatric brain tumors, including glioblastoma, diffuse midline gliomas, and medulloblastomas that continue to portend a grim prognosis. We describe insights gained from different preclinical models using CAR T therapy against various brain tumors and results gathered from ongoing clinical trials. Furthermore, we outline the challenges limiting CAR T therapy success against brain tumors and summarize advancements made to overcome these obstacles.
Collapse
Affiliation(s)
- Grace Guzman
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Megan R Reed
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
57
|
Tręda C, Włodarczyk A, Pacholczyk M, Rutkowska A, Stoczyńska-Fidelus E, Kierasińska A, Rieske P. Increased EGFRvIII Epitope Accessibility after Tyrosine Kinase Inhibitor Treatment of Glioblastoma Cells Creates More Opportunities for Immunotherapy. Int J Mol Sci 2023; 24:4350. [PMID: 36901782 PMCID: PMC10001577 DOI: 10.3390/ijms24054350] [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: 11/11/2022] [Revised: 02/10/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
The number of glioblastoma (GB) cases is increasing every year, and the currently available therapies remain ineffective. A prospective antigen for GB therapy is EGFRvIII, an EGFR deletion mutant containing a unique epitope that is recognized by the L8A4 antibody used in CAR-T (chimeric antigen receptor T cell) therapy. In this study, we observed that the concomitant use of L8A4 with particular tyrosine kinase inhibitors (TKIs) does not impede the interaction between L8A4 and EGFRvIII; moreover, in this case, the stabilization of formed dimers results in increased epitope display. Unlike in wild-type EGFR, a free cysteine at position 16 (C16) is exposed in the extracellular structure of EGFRvIII monomers, leading to covalent dimer formation in the region of L8A4-EGFRvIII mutual interaction. Following in silico analysis of cysteines possibly involved in covalent homodimerization, we prepared constructs containing cysteine-serine substitutions of EGFRvIII in adjacent regions. We found that the extracellular part of EGFRvIII possesses plasticity in the formation of disulfide bridges within EGFRvIII monomers and dimers due to the engagement of cysteines other than C16. Our results suggest that the EGFRvIII-specific L8A4 antibody recognizes both EGFRvIII monomers and covalent dimers, regardless of the cysteine bridging structure. To summarize, immunotherapy based on the L8A4 antibody, including CAR-T combined with TKIs, can potentially increase the chances of success in anti-GB therapy.
Collapse
Affiliation(s)
- Cezary Tręda
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| | - Aneta Włodarczyk
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| | - Marcin Pacholczyk
- Department of Research and Development, Celther Polska Ltd., Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - Adrianna Rutkowska
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| | - Ewelina Stoczyńska-Fidelus
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| | - Amelia Kierasińska
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| |
Collapse
|
58
|
Hamad A, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recent Developments in Glioblastoma Therapy: Oncolytic Viruses and Emerging Future Strategies. Viruses 2023; 15:547. [PMID: 36851761 PMCID: PMC9958853 DOI: 10.3390/v15020547] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma is the most aggressive form of malignant brain tumor. Standard treatment protocols and traditional immunotherapy are poorly effective as they do not significantly increase the long-term survival of glioblastoma patients. Oncolytic viruses (OVs) may be an effective alternative approach. Combining OVs with some modern treatment options may also provide significant benefits for glioblastoma patients. Here we review virotherapy for glioblastomas and describe several OVs and their combination with other therapies. The personalized use of OVs and their combination with other treatment options would become a significant area of research aiming to develop the most effective treatment regimens for glioblastomas.
Collapse
Affiliation(s)
- Azzam Hamad
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
59
|
Liang J, Fang D, Gumin J, Najem H, Sooreshjani M, Song R, Sabbagh A, Kong LY, Duffy J, Balyasnikova IV, Pollack SM, Puduvalli VK, Heimberger AB. A Case Study of Chimeric Antigen Receptor T Cell Function: Donor Therapeutic Differences in Activity and Modulation with Verteporfin. Cancers (Basel) 2023; 15:1085. [PMID: 36831427 PMCID: PMC9953964 DOI: 10.3390/cancers15041085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cells have recently been demonstrated to extract and express cognate tumor antigens through trogocytosis. This process may contribute to tumor antigen escape, T cell exhaustion, and fratricide, which plays a central role in CAR dysfunction. We sought to evaluate the importance of this effect in epidermal growth factor receptor variant III (EGFRvIII) specific CAR T cells targeting glioma. METHODS EGFRvIII-specific CAR T cells were generated from various donors and analyzed for cytotoxicity, trogocytosis, and in vivo therapeutic activity against intracranial glioma. Tumor autophagy resulting from CAR T cell activity was evaluated in combination with an autophagy inducer (verteporfin) or inhibitor (bafilomycin A1). RESULTS CAR T cell products derived from different donors induced markedly divergent levels of trogocytosis of tumor antigen as well as PD-L1 upon engaging target tumor cells correlating with variability in efficacy in mice. Pharmacological facilitation of CAR induced-autophagy with verteporfin inhibits trogocytic expression of tumor antigen on CARs and increases CAR persistence and efficacy in mice. CONCLUSION These data propose CAR-induced autophagy as a mechanism counteracting CAR-induced trogocytosis and provide a new strategy to innovate high-performance CARs through pharmacological facilitation of T cell-induced tumor death.
Collapse
Affiliation(s)
- Jiyong Liang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dexing Fang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hinda Najem
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Moloud Sooreshjani
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Renduo Song
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aria Sabbagh
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ling-Yuan Kong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joseph Duffy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Irina V. Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Seth M. Pollack
- Department of Cancer Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Vinay K. Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurosurgery, Northwestern University, Simpson Querrey Biomedical Research Center, 303 E. Superior Street, 6-516, Chicago, IL 60611, USA
| |
Collapse
|
60
|
CAR-T-Derived Extracellular Vesicles: A Promising Development of CAR-T Anti-Tumor Therapy. Cancers (Basel) 2023; 15:cancers15041052. [PMID: 36831396 PMCID: PMC9954490 DOI: 10.3390/cancers15041052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/26/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogenous population of plasma membrane-surrounded particles that are released in the extracellular milieu by almost all types of living cells. EVs are key players in intercellular crosstalk, both locally and systemically, given that they deliver their cargoes (consisting of proteins, lipids, mRNAs, miRNAs, and DNA fragments) to target cells, crossing biological barriers. Those mechanisms further trigger a wide range of biological responses. Interestingly, EV phenotypes and cargoes and, therefore, their functions, stem from their specific parental cells. For these reasons, EVs have been proposed as promising candidates for EV-based, cell-free therapies. One of the new frontiers of cell-based immunotherapy for the fight against refractory neoplastic diseases is represented by genetically engineered chimeric antigen receptor T (CAR-T) lymphocytes, which in recent years have demonstrated their effectiveness by reaching commercialization and clinical application for some neoplastic diseases. CAR-T-derived EVs represent a recent promising development of CAR-T immunotherapy approaches. This crosscutting innovative strategy is designed to exploit the advantages of genetically engineered cell-based immunotherapy together with those of cell-free EVs, which in principle might be safer and more efficient in crossing biological and tumor-associated barriers. In this review, we underlined the potential of CAR-T-derived EVs as therapeutic agents in tumors.
Collapse
|
61
|
Huang Z, Dewanjee S, Chakraborty P, Jha NK, Dey A, Gangopadhyay M, Chen XY, Wang J, Jha SK. CAR T cells: engineered immune cells to treat brain cancers and beyond. Mol Cancer 2023; 22:22. [PMID: 36721153 PMCID: PMC9890802 DOI: 10.1186/s12943-022-01712-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/29/2022] [Indexed: 02/01/2023] Open
Abstract
Malignant brain tumors rank among the most challenging type of malignancies to manage. The current treatment protocol commonly entails surgery followed by radiotherapy and/or chemotherapy, however, the median patient survival rate is poor. Recent developments in immunotherapy for a variety of tumor types spark optimism that immunological strategies may help patients with brain cancer. Chimeric antigen receptor (CAR) T cells exploit the tumor-targeting specificity of antibodies or receptor ligands to direct the cytolytic capacity of T cells. Several molecules have been discovered as potential targets for immunotherapy-based targeting, including but not limited to EGFRvIII, IL13Rα2, and HER2. The outstanding clinical responses to CAR T cell-based treatments in patients with hematological malignancies have generated interest in using this approach to treat solid tumors. Research results to date support the astounding clinical response rates of CD19-targeted CAR T cells, early clinical experiences in brain tumors demonstrating safety and evidence for disease-modifying activity, and the promise for further advances to ultimately assist patients clinically. However, several variable factors seem to slow down the progress rate regarding treating brain cancers utilizing CAR T cells. The current study offers a thorough analysis of CAR T cells' promise in treating brain cancer, including design and delivery considerations, current strides in clinical and preclinical research, issues encountered, and potential solutions.
Collapse
Affiliation(s)
- Zoufang Huang
- grid.452437.3Department of Hematology, Ganzhou Key Laboratory of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Saikat Dewanjee
- grid.216499.10000 0001 0722 3459Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Pratik Chakraborty
- grid.216499.10000 0001 0722 3459Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032 India
| | - Niraj Kumar Jha
- grid.412552.50000 0004 1764 278XDepartment of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh 201310 India
| | - Abhijit Dey
- grid.412537.60000 0004 1768 2925Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, West Bengal 700032 India
| | - Moumita Gangopadhyay
- grid.502979.00000 0004 6087 8632Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat, Kolkata, West Bengal 700126 India
| | - Xuan-Yu Chen
- grid.264091.80000 0001 1954 7928Institute for Biotechnology, St. John’s University, Queens, New York, 11439 USA
| | - Jian Wang
- Department of Radiotherapy, the Affiliated Jiangyin People’s Hospital of Nantong University, Jiangyin, 214400 China
| | - Saurabh Kumar Jha
- grid.412552.50000 0004 1764 278XDepartment of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh 201310 India ,grid.448792.40000 0004 4678 9721Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413 India ,grid.449906.60000 0004 4659 5193Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007 India
| |
Collapse
|
62
|
Yan T, Zhu L, Chen J. Current advances and challenges in CAR T-Cell therapy for solid tumors: tumor-associated antigens and the tumor microenvironment. Exp Hematol Oncol 2023; 12:14. [PMID: 36707873 PMCID: PMC9883880 DOI: 10.1186/s40164-023-00373-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
The past decade has witnessed ongoing progress in immune therapy to ameliorate human health. As an emerging technique, chimeric antigen receptor (CAR) T-cell therapy has the advantages of specific killing of cancer cells, a high remission rate of cancer-induced symptoms, rapid tumor eradication, and long-lasting tumor immunity, opening a new window for tumor treatment. However, challenges remain in CAR T-cell therapy for solid tumors due to target diversity, tumor heterogeneity, and the complex microenvironment. In this review, we have outlined the development of the CAR T-cell technique, summarized the current advances in tumor-associated antigens (TAAs), and highlighted the importance of tumor-specific antigens (TSAs) or neoantigens for solid tumors. We also addressed the challenge of the TAA binding domain in CARs to overcome off-tumor toxicity. Moreover, we illustrated the dominant tumor microenvironment (TME)-induced challenges and new strategies based on TME-associated antigens (TMAs) for solid tumor CAR T-cell therapy.
Collapse
Affiliation(s)
- Ting Yan
- grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| | - Lingfeng Zhu
- grid.443397.e0000 0004 0368 7493Department of Urology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| | - Jin Chen
- grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China ,grid.443397.e0000 0004 0368 7493Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| |
Collapse
|
63
|
The Tumor Immune Microenvironment in Primary CNS Neoplasms: A Review of Current Knowledge and Therapeutic Approaches. Int J Mol Sci 2023; 24:ijms24032020. [PMID: 36768342 PMCID: PMC9917056 DOI: 10.3390/ijms24032020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Primary CNS neoplasms are responsible for considerable mortality and morbidity, and many therapies directed at primary brain tumors have proven unsuccessful despite their success in preclinical studies. Recently, the tumor immune microenvironment has emerged as a critical aspect of primary CNS neoplasms that may affect their malignancy, prognosis, and response to therapy across patients and tumor grades. This review covers the tumor microenvironment of various primary CNS neoplasms, with a focus on glioblastoma and meningioma. Additionally, current therapeutic strategies based on elements of the tumor microenvironment, including checkpoint inhibitor therapy and immunotherapeutic vaccines, are discussed.
Collapse
|
64
|
Wang G, Zhang Z, Zhong K, Wang Z, Yang N, Tang X, Li H, Lu Q, Wu Z, Yuan B, Zheng M, Cheng P, Tong A, Zhou L. CXCL11-armed oncolytic adenoviruses enhance CAR-T cell therapeutic efficacy and reprogram tumor microenvironment in glioblastoma. Mol Ther 2023; 31:134-153. [PMID: 36056553 PMCID: PMC9840126 DOI: 10.1016/j.ymthe.2022.08.021] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/15/2022] [Accepted: 08/26/2022] [Indexed: 01/28/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary malignant brain cancer and urgently requires effective treatments. Chimeric antigen receptor T (CAR-T) cell therapy offers a potential treatment method, but it is often hindered by poor infiltration of CAR-T cells in tumors and highly immunosuppressive tumor microenvironment (TME). Here, we armed an oncolytic adenovirus (oAds) with a chemokine CXCL11 to increase the infiltration of CAR-T cells and reprogram the immunosuppressive TME, thus improving its therapeutic efficacy. In both immunodeficient and immunocompetent orthotopic GBM mice models, we showed that B7H3-targeted CAR-T cells alone failed to inhibit GBM growth but, when combined with the intratumoral administration of CXCL11-armed oAd, it achieved a durable antitumor response. Besides, oAd-CXCL11 had a potent antitumor effect and reprogramed the immunosuppressive TME in GL261 GBM models, in which increased infiltration of CD8+ T lymphocytes, natural killer (NK) cells, and M1-polarized macrophages, while decreased proportions of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and M2-polarized macrophages were observed. Furthermore, the antitumor effect of the oAd-CXCL11 was CD8+ T cell dependent. Our findings thus revealed that CXCL11-armed oAd can improve immune-virotherapy and can be a promising adjuvant of CAR-T therapy for GBM.
Collapse
Affiliation(s)
- Guoqing Wang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Kunhong Zhong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zeng Wang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Nian Yang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xin Tang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Hexian Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qizhong Lu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhiguo Wu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Boyang Yuan
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Ping Cheng
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
65
|
Swan SL, Mehta N, Ilich E, Shen SH, Wilkinson DS, Anderson AR, Segura T, Sanchez-Perez L, Sampson JH, Bellamkonda RV. IL7 and IL7 Flt3L co-expressing CAR T cells improve therapeutic efficacy in mouse EGFRvIII heterogeneous glioblastoma. Front Immunol 2023; 14:1085547. [PMID: 36817432 PMCID: PMC9936235 DOI: 10.3389/fimmu.2023.1085547] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy in glioblastoma faces many challenges including insufficient CAR T cell abundance and antigen-negative tumor cells evading targeting. Unfortunately, preclinical studies evaluating CAR T cells in glioblastoma focus on tumor models that express a single antigen, use immunocompromised animals, and/or pre-treat with lymphodepleting agents. While lymphodepletion enhances CAR T cell efficacy, it diminishes the endogenous immune system that has the potential for tumor eradication. Here, we engineered CAR T cells to express IL7 and/or Flt3L in 50% EGFRvIII-positive and -negative orthotopic tumors pre-conditioned with non-lymphodepleting irradiation. IL7 and IL7 Flt3L CAR T cells increased intratumoral CAR T cell abundance seven days after treatment. IL7 co-expression with Flt3L modestly increased conventional dendritic cells as well as the CD103+XCR1+ population known to have migratory and antigen cross-presenting capabilities. Treatment with IL7 or IL7 Flt3L CAR T cells improved overall survival to 67% and 50%, respectively, compared to 9% survival with conventional or Flt3L CAR T cells. We concluded that CAR T cells modified to express IL7 enhanced CAR T cell abundance and improved overall survival in EGFRvIII heterogeneous tumors pre-conditioned with non-lymphodepleting irradiation. Potentially IL7 or IL7 Flt3L CAR T cells can provide new opportunities to combine CAR T cells with other immunotherapies for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Sheridan L Swan
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Nalini Mehta
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Ekaterina Ilich
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Steven H Shen
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Daniel S Wilkinson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Alexa R Anderson
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| | - Tatiana Segura
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States.,Clinical Science Departments of Neurology and Dermatology, Duke University, Durham, NC, United States
| | - Luis Sanchez-Perez
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States.,The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC, United States.,Department of Pathology, Duke University Medical Center, Durham, NC, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, NC, United States
| | - Ravi V Bellamkonda
- Department of Biology, Emory University, Atlanta, GA, United States.,Wallace H. Coulter Department of Biomedical Engineering, Emory University, Atlanta, GA, United States
| |
Collapse
|
66
|
Saleh HA, Mitwasi N, Ullrich M, Kubeil M, Toussaint M, Deuther-Conrad W, Neuber C, Arndt C, R. Loureiro L, Kegler A, González Soto KE, Belter B, Rössig C, Pietzsch J, Frenz M, Bachmann M, Feldmann A. Specific and safe targeting of glioblastoma using switchable and logic-gated RevCAR T cells. Front Immunol 2023; 14:1166169. [PMID: 37122703 PMCID: PMC10145173 DOI: 10.3389/fimmu.2023.1166169] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Glioblastoma (GBM) is still an incurable tumor that is associated with high recurrence rate and poor survival despite the current treatment regimes. With the urgent need for novel therapeutic strategies, immunotherapies, especially chimeric antigen receptor (CAR)-expressing T cells, represent a promising approach for specific and effective targeting of GBM. However, CAR T cells can be associated with serious side effects. To overcome such limitation, we applied our switchable RevCAR system to target both the epidermal growth factor receptor (EGFR) and the disialoganglioside GD2, which are expressed in GBM. The RevCAR system is a modular platform that enables controllability, improves safety, specificity and flexibility. Briefly, it consists of RevCAR T cells having a peptide epitope as extracellular domain, and a bispecific target module (RevTM). The RevTM acts as a switch key that recognizes the RevCAR epitope and the tumor-associated antigen, and thereby activating the RevCAR T cells to kill the tumor cells. However, in the absence of the RevTM, the RevCAR T cells are switched off. In this study, we show that the novel EGFR/GD2-specific RevTMs can selectively activate RevCAR T cells to kill GBM cells. Moreover, we show that gated targeting of GBM is possible with our Dual-RevCAR T cells, which have their internal activation and co-stimulatory domains separated into two receptors. Therefore, a full activation of Dual-RevCAR T cells can only be achieved when both receptors recognize EGFR and GD2 simultaneously via RevTMs, leading to a significant killing of GBM cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Haidy A. Saleh
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Nicola Mitwasi
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Manja Kubeil
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Magali Toussaint
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Arndt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Faculty of Medicine Carl Gustav Carus, Mildred Scheel Early Career Center, Technische Universität Dresden, Dresden, Germany
| | - Liliana R. Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Alexandra Kegler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | | | - Birgit Belter
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Claudia Rössig
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Münster, Münster, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - Marcus Frenz
- Faculty Informatik and Wirtschaftsinformatik, Provadis School of International Management and Technology AG, Frankfurt, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site, Dresden, Germany
- *Correspondence: Michael Bachmann,
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site, Dresden, Germany
| |
Collapse
|
67
|
Abdulmajeed F, Hamandi M, Malaiyandi D, Shutter L. Neurocritical Care in the General Intensive Care Unit. Crit Care Clin 2023; 39:153-169. [DOI: 10.1016/j.ccc.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
68
|
Song EZ, Wang X, Philipson BI, Zhang Q, Thokala R, Zhang L, Assenmacher CA, Binder ZA, Ming GL, O’Rourke DM, Song H, Milone MC. The IAP antagonist birinapant enhances chimeric antigen receptor T cell therapy for glioblastoma by overcoming antigen heterogeneity. Mol Ther Oncolytics 2022; 27:288-304. [PMID: 36458202 PMCID: PMC9707011 DOI: 10.1016/j.omto.2022.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
Antigen heterogeneity that results in tumor antigenic escape is one of the major obstacles to successful chimeric antigen receptor (CAR) T cell therapies in solid tumors including glioblastoma multiforme (GBM). To address this issue and improve the efficacy of CAR T cell therapy for GBM, we developed an approach that combines CAR T cells with inhibitor of apoptosis protein (IAP) antagonists, a new class of small molecules that mediate the degradation of IAPs, to treat GBM. Here, we demonstrated that the IAP antagonist birinapant could sensitize GBM cell lines and patient-derived primary GBM organoids to apoptosis induced by CAR T cell-derived cytokines, such as tumor necrosis factor. Therefore, birinapant could enhance CAR T cell-mediated bystander death of antigen-negative GBM cells, thus preventing tumor antigenic escape in antigen-heterogeneous tumor models in vitro and in vivo. In addition, birinapant could promote the activation of NF-κB signaling pathways in antigen-stimulated CAR T cells, and with a birinapant-resistant tumor model we showed that birinapant had no deleterious effect on CAR T cell functions in vitro and in vivo. Overall, we demonstrated the potential of combining the IAP antagonist birinapant with CAR T cells as a novel and feasible approach to overcoming tumor antigen heterogeneity and enhancing CAR T cell therapy for GBM.
Collapse
Affiliation(s)
- Edward Z. Song
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Xin Wang
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamin I. Philipson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Qian Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Radhika Thokala
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| | - Logan Zhang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles-Antoine Assenmacher
- Comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zev A. Binder
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-li Ming
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donald M. O’Rourke
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael C. Milone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, PCAM SPE 8-101, Philadelphia, PA 19104, USA
| |
Collapse
|
69
|
Enam SF, Kilic CY, Huang J, Kang BJ, Chen R, Tribble CS, Ilich E, Betancur MI, Blocker SJ, Owen SJ, Buckley AF, Lyon JG, Bellamkonda RV. Cytostatic hypothermia and its impact on glioblastoma and survival. SCIENCE ADVANCES 2022; 8:eabq4882. [PMID: 36427309 PMCID: PMC9699673 DOI: 10.1126/sciadv.abq4882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
Patients with glioblastoma (GBM) have limited options and require novel approaches to treatment. Here, we studied and deployed nonfreezing "cytostatic" hypothermia to stunt GBM growth. This growth-halting method contrasts with ablative, cryogenic hypothermia that kills both neoplastic and infiltrated healthy tissue. We investigated degrees of hypothermia in vitro and identified a cytostatic window of 20° to 25°C. For some lines, 18 hours/day of cytostatic hypothermia was sufficient to halt division in vitro. Next, we fabricated an experimental tool to test local cytostatic hypothermia in two rodent GBM models. Hypothermia more than doubled median survival, and all rats that successfully received cytostatic hypothermia survived their study period. Unlike targeted therapeutics that are successful in preclinical models but fail in clinical trials, cytostatic hypothermia leverages fundamental physics that influences biology broadly. It is a previously unexplored approach that could provide an additional option to patients with GBM by halting tumor growth.
Collapse
Affiliation(s)
- Syed Faaiz Enam
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Cem Y. Kilic
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Jianxi Huang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Brian J. Kang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Reed Chen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Connor S. Tribble
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Ekaterina Ilich
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Martha I. Betancur
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Stephanie J. Blocker
- Department of Radiology, Center for In Vivo Microscopy, Duke University, Durham, NC 27705, USA
| | - Steven J. Owen
- Bio-medical Machine Shop, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
| | - Anne F. Buckley
- Department of Pathology, School of Medicine, Duke University, Durham, NC 27705, USA
| | - Johnathan G. Lyon
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
- Department of Biology, Emory University, Atlanta, GA 30332, USA
| | - Ravi V. Bellamkonda
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27705, USA
- Department of Biology, Emory University, Atlanta, GA 30332, USA
| |
Collapse
|
70
|
Zhao S, Li J, Zhang H, Qi L, Du Y, Kogiso M, Braun FK, Xiao S, Huang Y, Li J, Teo WY, Lindsay H, Baxter P, Su JMF, Adesina A, Laczik M, Genevini P, Veillard AC, Schvartzman S, Berguet G, Ding SR, Du L, Stephan C, Yang J, Davies PJA, Lu X, Chintagumpala M, Parsons DW, Perlaky L, Xia YF, Man TK, Huang Y, Sun D, Li XN. Epigenetic Alterations of Repeated Relapses in Patient-matched Childhood Ependymomas. Nat Commun 2022; 13:6689. [PMID: 36335125 PMCID: PMC9637194 DOI: 10.1038/s41467-022-34514-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022] Open
Abstract
Recurrence is frequent in pediatric ependymoma (EPN). Our longitudinal integrated analysis of 30 patient-matched repeated relapses (3.67 ± 1.76 times) over 13 years (5.8 ± 3.8) reveals stable molecular subtypes (RELA and PFA) and convergent DNA methylation reprogramming during serial relapses accompanied by increased orthotopic patient derived xenograft (PDX) (13/27) formation in the late recurrences. A set of differentially methylated CpGs (DMCs) and DNA methylation regions (DMRs) are found to persist in primary and relapse tumors (potential driver DMCs) and are acquired exclusively in the relapses (potential booster DMCs). Integrating with RNAseq reveals differentially expressed genes regulated by potential driver DMRs (CACNA1H, SLC12A7, RARA in RELA and HSPB8, GMPR, ITGB4 in PFA) and potential booster DMRs (PLEKHG1 in RELA and NOTCH, EPHA2, SUFU, FOXJ1 in PFA tumors). DMCs predicators of relapse are also identified in the primary tumors. This study provides a high-resolution epigenetic roadmap of serial EPN relapses and 13 orthotopic PDX models to facilitate biological and preclinical studies.
Collapse
Affiliation(s)
- Sibo Zhao
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.413584.f0000 0004 0383 5679Jane and John Justin Neurosciences Center, Cook Children’s Medical Center, Fort Worth, TX 76104 USA ,grid.413584.f0000 0004 0383 5679Hematology and Oncology Center, Cook Children’s Medical Center, Fort Worth, TX 76104 USA
| | - Jia Li
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA ,grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA ,grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University; and Guangzhou Laboratory, Bioland, 510120 Guangzhou, Guangdong P. R. China
| | - Huiyuan Zhang
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Lin Qi
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Yuchen Du
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Mari Kogiso
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Frank K. Braun
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Sophie Xiao
- grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Yulun Huang
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.263761.70000 0001 0198 0694Department of Neurosurgery and Brain and Nerve Research Laboratory, the First Affiliated Hospital, and Department of Neurosurgery, Dushu Lake Hospital, Suzhou Medical College, Soochow University, 215007 Suzhou, P. R. China
| | - Jianfang Li
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Wan-Yee Teo
- grid.410724.40000 0004 0620 9745Humphrey Oei Institute of Cancer Research, National Cancer Center Singapore, Singapore, 169610 Singapore ,grid.428397.30000 0004 0385 0924Cancer and Stem Cell Biology Program, Duke-NUS Medical School Singapore, Singapore, Singapore ,grid.414963.d0000 0000 8958 3388KK Women’s & Children’s Hospital Singapore, Singapore, Singapore ,grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Holly Lindsay
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Patricia Baxter
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Jack M. F. Su
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Adekunle Adesina
- grid.39382.330000 0001 2160 926XDepartment of Pathology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Miklós Laczik
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Paola Genevini
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | | | - Sol Schvartzman
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Geoffrey Berguet
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Shi-Rong Ding
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Department of Radiation, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong P. R. China
| | - Liping Du
- grid.16753.360000 0001 2299 3507Clinical Cytogenetic Laboratory, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Clifford Stephan
- grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA
| | - Jianhua Yang
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Peter J. A. Davies
- grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA
| | - Xinyan Lu
- grid.16753.360000 0001 2299 3507Clinical Cytogenetic Laboratory, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Murali Chintagumpala
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Donald William Parsons
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Laszlo Perlaky
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yun-Fei Xia
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Department of Radiation, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong P. R. China
| | - Tsz-Kwong Man
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yun Huang
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Deqiang Sun
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Xiao-Nan Li
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
71
|
Abstract
CAR T-cell therapy has transformed the treatment of hematological malignancies of the B cell lineage. However, the quest to fulfil the same promise for solid tumors is still in its infancy. This review summarizes some of the challenges that the field is trying to overcome for effective treatment of human carcinomas, including tumor heterogeneity, the paucity of truly tumor-specific targets, immunosuppression and metabolic restrictions at solid tumor beds, and defective T-cell trafficking. All these barriers are being currently investigated and, in some cases, targeted, by multiple independent groups. With clinical interventions against multiple human malignancies and different platforms under accelerated clinical development, the next few years will see an array of cellular therapies, including CAR T-cells, progressively becoming routine interventions to eliminate currently incurable diseases, as it happened with some hematological malignancies.
Collapse
Affiliation(s)
- Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Department of Gynecologic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Jose A Guevara-Patino
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
72
|
Mamun AA, Uddin MS, Perveen A, Jha NK, Alghamdi BS, Jeandet P, Zhang HJ, Ashraf GM. Inflammation-targeted nanomedicine against brain cancer: From design strategies to future developments. Semin Cancer Biol 2022; 86:101-116. [PMID: 36084815 DOI: 10.1016/j.semcancer.2022.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 08/08/2022] [Accepted: 08/21/2022] [Indexed: 02/07/2023]
Abstract
Brain cancer is an aggressive type of cancer with poor prognosis. While the immune system protects against cancer in the early stages, the tumor exploits the healing arm of inflammatory reactions to accelerate its growth and spread. Various immune cells penetrate the developing tumor region, establishing a pro-inflammatory tumor milieu. Additionally, tumor cells may release chemokines and cytokines to attract immune cells and promote cancer growth. Inflammation and its associated mechanisms in the progression of cancer have been extensively studied in the majority of solid tumors, especially brain tumors. However, treatment of the malignant brain cancer is hindered by several obstacles, such as the blood-brain barrier, transportation inside the brain interstitium, inflammatory mediators that promote tumor growth and invasiveness, complications in administering therapies to tumor cells specifically, the highly invasive nature of gliomas, and the resistance to drugs. To resolve these obstacles, nanomedicine could be a potential strategy that has facilitated advancements in diagnosing and treating brain cancer. Due to the numerous benefits provided by their small size and other features, nanoparticles have been a prominent focus of research in the drug-delivery field. The purpose of this article is to discuss the role of inflammatory mediators and signaling pathways in brain cancer as well as the recent advances in understanding the nano-carrier approaches for enhancing drug delivery to the brain in the treatment of brain cancer.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region of China
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences, Uttaranchal University, Dehradun 248007, India
| | - Badrah S Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Philippe Jeandet
- University of Reims Champagne-Ardenne, Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, PO Box 1039, 51687 Reims Cedex 2, France
| | - Hong-Jie Zhang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong Special Administrative Region of China
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
73
|
Karami Fath M, Babakhaniyan K, Anjomrooz M, Jalalifar M, Alizadeh SD, Pourghasem Z, Abbasi Oshagh P, Azargoonjahromi A, Almasi F, Manzoor HZ, Khalesi B, Pourzardosht N, Khalili S, Payandeh Z. Recent Advances in Glioma Cancer Treatment: Conventional and Epigenetic Realms. Vaccines (Basel) 2022; 10:1448. [PMID: 36146527 PMCID: PMC9501259 DOI: 10.3390/vaccines10091448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/14/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most typical and aggressive form of primary brain tumor in adults, with a poor prognosis. Successful glioma treatment is hampered by ineffective medication distribution across the blood-brain barrier (BBB) and the emergence of drug resistance. Although a few FDA-approved multimodal treatments are available for glioblastoma, most patients still have poor prognoses. Targeting epigenetic variables, immunotherapy, gene therapy, and different vaccine- and peptide-based treatments are some innovative approaches to improve anti-glioma treatment efficacy. Following the identification of lymphatics in the central nervous system, immunotherapy offers a potential method with the potency to permeate the blood-brain barrier. This review will discuss the rationale, tactics, benefits, and drawbacks of current glioma therapy options in clinical and preclinical investigations.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran 1571914911, Iran
| | - Kimiya Babakhaniyan
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran 1996713883, Iran
| | - Mehran Anjomrooz
- Department of Radiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1411713135, Iran
| | | | | | - Zeinab Pourghasem
- Department of Microbiology, Islamic Azad University of Lahijan, Gilan 4416939515, Iran
| | - Parisa Abbasi Oshagh
- Department of Biology, Faculty of Basic Sciences, Malayer University, Malayer 6571995863, Iran
| | - Ali Azargoonjahromi
- Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz 7417773539, Iran
| | - Faezeh Almasi
- Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 1411734115, Iran
| | - Hafza Zahira Manzoor
- Experimental and Translational Medicine, University of Insubria, Via jean Henry Dunant 3, 21100 Varese, Italy
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3197619751, Iran
| | - Navid Pourzardosht
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht 4193713111, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran 1678815811, Iran
| | - Zahra Payandeh
- Department of Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, SE-17177 Stockholm, Sweden
| |
Collapse
|
74
|
Wang G, Wang W. Advanced Cell Therapies for Glioblastoma. Front Immunol 2022; 13:904133. [PMID: 36052072 PMCID: PMC9425637 DOI: 10.3389/fimmu.2022.904133] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
The sheer ubiquity of Gioblastoma (GBM) cases would lead you to believe that there should have been many opportunities for the discovery of treatments to successfully render it into remission. Unfortunately, its persistent commonality is due in large part to the fact that it is the most treatment-resistant tumors in adults. That completely changes the treatment plan of attack. Long established and accepted treatment therapies such as surgical resection, radiation, and aggressive chemotherapy, (and any combination thereof) have only confirmed that the disease lives up to its treatment-resistant reputation. To add to the seemingly insurmountable task of finding a cure, GBM has also proven to be a very stubborn and formidable opponent to newer immunotherapies. Across the board, regardless of the therapy combination, the five-year survival rate of GBM patients is still very poor at a heartbreaking 5.6%. Obviously, the present situation cannot be tolerated or deemed acceptable. The grave situation calls for researchers to be more innovative and find more efficient strategies to discover new and successful strategies to treat GBM. Inspired by researchers worldwide attempting to control GBM, we provide in this review a comprehensive overview of the many diverse cell therapies currently being used to treat GBM. An overview of the treatments include: CAR T cells, CAR NK cells, gamma-delta T cells, NKT cells, dendritic cells, macrophages, as well stem cell-based strategies. To give you the complete picture, we will discuss the efficacy, safety, and developmental stages, the mechanisms of action and the challenges of each of these therapies and detail their potential to be the next-generation immunotherapeutic to eliminate this dreadful disease.
Collapse
Affiliation(s)
- Guangwen Wang
- BlueRock Therapeutics, Department of Process Development, Cambridge, MA, United States
- *Correspondence: Wenshi Wang, ; Guangwen Wang,
| | - Wenshi Wang
- Metagenomi Inc., Department of Cell Therapy, Emeryville, CA, United States
- *Correspondence: Wenshi Wang, ; Guangwen Wang,
| |
Collapse
|
75
|
Asija S, Chatterjee A, Yadav S, Chekuri G, Karulkar A, Jaiswal AK, Goda JS, Purwar R. Combinatorial approaches to effective therapy in glioblastoma (GBM): Current status and what the future holds. Int Rev Immunol 2022; 41:582-605. [PMID: 35938932 DOI: 10.1080/08830185.2022.2101647] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
The aggressive and recurrent nature of glioblastoma is multifactorial and has been attributed to its biological heterogeneity, dysfunctional metabolic signaling pathways, rigid blood-brain barrier, inherent resistance to standard therapy due to the stemness property of the gliomas cells, immunosuppressive tumor microenvironment, hypoxia and neoangiogenesis which are very well orchestrated and create the tumor's own highly pro-tumorigenic milieu. Once the relay of events starts amongst these components, eventually it becomes difficult to control the cascade using only the balanced contemporary care of treatment consisting of maximal resection, radiotherapy and chemotherapy with temozolamide. Over the past few decades, implementation of contemporary treatment modalities has shown benefit to some extent, but no significant overall survival benefit is achieved. Therefore, there is an unmet need for advanced multifaceted combinatorial strategies. Recent advances in molecular biology, development of innovative therapeutics and novel delivery platforms over the years has resulted in a paradigm shift in gliomas therapeutics. Decades of research has led to emergence of several treatment molecules, including immunotherapies such as immune checkpoint blockade, oncolytic virotherapy, adoptive cell therapy, nanoparticles, CED and BNCT, each with the unique proficiency to overcome the mentioned challenges, present research. Recent years are seeing innovative combinatorial strategies to overcome the multifactorial resistance put forth by the GBM cell and its TME. This review discusses the contemporary and the investigational combinatorial strategies being employed to treat GBM and summarizes the evidence accumulated till date.
Collapse
Affiliation(s)
- Sweety Asija
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sandhya Yadav
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Godhanjali Chekuri
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Atharva Karulkar
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| | - Ankesh Kumar Jaiswal
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| | - Jayant S Goda
- Department of Radiation Oncology, Tata Memorial Center, Mumbai, Maharashtra, India.,Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Rahul Purwar
- Department of Biosciences & Bioengineering, Indian Institute of Technology, Mumbai, India
| |
Collapse
|
76
|
Sun J, Li X, Chen P, Gao Y. From Anti-HER-2 to Anti-HER-2-CAR-T Cells: An Evolutionary Immunotherapy Approach for Gastric Cancer. J Inflamm Res 2022; 15:4061-4085. [PMID: 35873388 PMCID: PMC9304417 DOI: 10.2147/jir.s368138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Current Therapeutic modalities provide no survival advantage to gastric cancer (GC) patients. Targeting the human epidermal growth factor receptor-2 (HER-2) is a viable therapeutic strategy against advanced HER-2 positive GC. Antibody-drug conjugates, small-molecule tyrosine kinase inhibitors (TKIs), and bispecific antibodies are emerging as novel drug forms that may abrogate the resistance to HER-2-specific drugs and monoclonal antibodies. Chimeric antigen receptor-modified T cells (CAR-T) targeting HER-2 have shown considerable therapeutic potential in GC and other solid tumors. However, due to the high heterogeneity along with the complex tumor microenvironment (TME) of GC that often leads to immune escape, the immunological treatment of GC still faces many challenges. Here, we reviewed and discussed the current progress in the research of anti-HER-2-CAR-T cell immunotherapy against GC.
Collapse
Affiliation(s)
- Jiangang Sun
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Xiaojing Li
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Peng Chen
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| | - Yongshun Gao
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, People's Republic of China
| |
Collapse
|
77
|
Site-Specific Considerations on Engineered T Cells for Malignant Gliomas. Biomedicines 2022; 10:biomedicines10071738. [PMID: 35885047 PMCID: PMC9312945 DOI: 10.3390/biomedicines10071738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy has revolutionized cancer treatment. Despite the recent advances in immunotherapeutic approaches for several tumor entities, limited response has been observed in malignant gliomas, including glioblastoma (GBM). Conversely, one of the emerging immunotherapeutic modalities is chimeric antigen receptors (CAR) T cell therapy, which demonstrated promising clinical responses in other solid tumors. Current pre-clinical and interventional clinical studies suggest improved efficacy when CAR-T cells are delivered locoregionally, rather than intravenously. In this review, we summarize possible CAR-T cell administration routes including locoregional therapy, systemic administration with and without focused ultrasound, direct intra-arterial drug delivery and nanoparticle-enhanced delivery in glioma. Moreover, we discuss published as well as ongoing and planned clinical trials involving CAR-T cell therapy in malignant glioma. With increasing neoadjuvant and/or adjuvant combinatorial immunotherapeutic concepts and modalities with specific modes of action for malignant glioma, selection of administration routes becomes increasingly important.
Collapse
|
78
|
Chen L, Chen F, Niu H, Li J, Pu Y, Yang C, Wang Y, Huang R, Li K, Lei Y, Huang Y. Chimeric Antigen Receptor (CAR)-T Cell Immunotherapy Against Thoracic Malignancies: Challenges and Opportunities. Front Immunol 2022; 13:871661. [PMID: 35911706 PMCID: PMC9334018 DOI: 10.3389/fimmu.2022.871661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Different from surgery, chemical therapy, radio-therapy and target therapy, Chimeric antigen receptor-modified T (CAR-T) cells, a novel adoptive immunotherapy strategy, have been used successfully against both hematological tumors and solid tumors. Although several problems have reduced engineered CAR-T cell therapeutic outcomes in clinical trials for the treatment of thoracic malignancies, including the lack of specific antigens, an immunosuppressive tumor microenvironment, a low level of CAR-T cell infiltration into tumor tissues, off-target toxicity, and other safety issues, CAR-T cell treatment is still full of bright future. In this review, we outline the basic structure and characteristics of CAR-T cells among different period, summarize the common tumor-associated antigens in clinical trials of CAR-T cell therapy for thoracic malignancies, and point out the current challenges and new strategies, aiming to provide new ideas and approaches for preclinical experiments and clinical trials of CAR-T cell therapy for thoracic malignancies.
Collapse
Affiliation(s)
- Long Chen
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Huatao Niu
- Department of Neurosurgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Jindan Li
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yongzhu Pu
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Conghui Yang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yue Wang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Rong Huang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Ke Li
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yujie Lei
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| |
Collapse
|
79
|
Guda MR, Tsung AJ, Asuthkar S, Velpula KK. Galectin-1 activates carbonic anhydrase IX and modulates glioma metabolism. Cell Death Dis 2022; 13:574. [PMID: 35773253 PMCID: PMC9247167 DOI: 10.1038/s41419-022-05024-z] [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: 02/09/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 01/21/2023]
Abstract
Galectins are a family of β-galactose-specific binding proteins residing within the cytosol or nucleus, with a highly conserved carbohydrate recognition domain across many species. Accumulating evidence shows that Galectin 1 (Gal-1) plays an essential role in cancer, and its expression correlates with tumor aggressiveness and progression. Our preliminary data showed Gal-1 promotes glioma stem cell (GSC) growth via increased Warburg effect. mRNA expression and clinical data were obtained from The Cancer Genome Atlas database. The immunoblot analysis conducted using our cohort of human glioblastoma patient specimens (hGBM), confirmed Gal-1 upregulation in GBM. GC/MS analysis to evaluate the effects of Gal-1 depletion showed elevated levels of α-ketoglutaric acid, and citric acid with a concomitant reduction in lactic acid levels. Using Biolog microplate-1 mitochondrial functional assay, we confirmed that the depletion of Gal-1 increases the expression levels of the enzymes from the TCA cycle, suggesting a reversal of the Warburg phenotype. Manipulation of Gal-1 using RNA interference showed reduced ATP, lactate levels, cell viability, colony-forming abilities, and increased expression levels of genes implicated in the induction of apoptosis. Gal-1 exerts its metabolic role via regulating the expression of carbonic anhydrase IX (CA-IX), a surrogate marker for hypoxia. CA-IX functions downstream to Gal-1, and co-immunoprecipitation experiments along with proximity ligation assays confirm that Gal-1 physically associates with CA-IX to regulate its expression. Further, silencing of Gal-1 in mice models showed reduced tumor burden and increased survival compared to the mice implanted with GSC controls. Further investigation of Gal-1 in GSC progression and metabolic reprogramming is warranted.
Collapse
Affiliation(s)
- Maheedhara R. Guda
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA
| | - Andrew J. Tsung
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,grid.430852.80000 0001 0741 4132Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,Illinois Neurological Institute, Peoria, IL USA
| | - Swapna Asuthkar
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA
| | - Kiran K. Velpula
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,grid.430852.80000 0001 0741 4132Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,grid.430852.80000 0001 0741 4132Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL USA
| |
Collapse
|
80
|
Rodríguez-Camacho A, Flores-Vázquez JG, Moscardini-Martelli J, Torres-Ríos JA, Olmos-Guzmán A, Ortiz-Arce CS, Cid-Sánchez DR, Pérez SR, Macías-González MDS, Hernández-Sánchez LC, Heredia-Gutiérrez JC, Contreras-Palafox GA, Suárez-Campos JDJE, Celis-López MÁ, Gutiérrez-Aceves GA, Moreno-Jiménez S. Glioblastoma Treatment: State-of-the-Art and Future Perspectives. Int J Mol Sci 2022; 23:ijms23137207. [PMID: 35806212 PMCID: PMC9267036 DOI: 10.3390/ijms23137207] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/09/2022] [Accepted: 06/25/2022] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Glioblastoma is the most frequent and lethal primary tumor of the central nervous system. Through many years, research has brought various advances in glioblastoma treatment. At this time, glioblastoma management is based on maximal safe surgical resection, radiotherapy, and chemotherapy with temozolomide. Recently, bevacizumab has been added to the treatment arsenal for the recurrent scenario. Nevertheless, patients with glioblastoma still have a poor prognosis. Therefore, many efforts are being made in different clinical research areas to find a new alternative to improve overall survival, free-progression survival, and life quality in glioblastoma patients. (2) Methods: Our objective is to recap the actual state-of-the-art in glioblastoma treatment, resume the actual research and future perspectives on immunotherapy, as well as the new synthetic molecules and natural compounds that represent potential future therapies at preclinical stages. (3) Conclusions: Despite the great efforts in therapeutic research, glioblastoma management has suffered minimal changes, and the prognosis remains poor. Combined therapeutic strategies and delivery methods, including immunotherapy, synthetic molecules, natural compounds, and glioblastoma stem cell inhibition, may potentiate the standard of care therapy and represent the next step in glioblastoma management research.
Collapse
Affiliation(s)
- Alejandro Rodríguez-Camacho
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - José Guillermo Flores-Vázquez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- Correspondence:
| | - Júlia Moscardini-Martelli
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Jorge Alejandro Torres-Ríos
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Alejandro Olmos-Guzmán
- Hospital de Especialidades No.1 Centro Médico Nacional del Bajío, León 37680, Mexico; (A.O.-G.); (C.S.O.-A.)
| | - Cindy Sharon Ortiz-Arce
- Hospital de Especialidades No.1 Centro Médico Nacional del Bajío, León 37680, Mexico; (A.O.-G.); (C.S.O.-A.)
| | - Dharely Raquel Cid-Sánchez
- Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (D.R.C.-S.); (S.R.P.)
| | - Samuel Rosales Pérez
- Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (D.R.C.-S.); (S.R.P.)
| | | | - Laura Crystell Hernández-Sánchez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Juan Carlos Heredia-Gutiérrez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Gabriel Alejandro Contreras-Palafox
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - José de Jesús Emilio Suárez-Campos
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Miguel Ángel Celis-López
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Guillermo Axayacalt Gutiérrez-Aceves
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Sergio Moreno-Jiménez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (A.R.-C.); (J.M.-M.); (J.A.T.-R.); (L.C.H.-S.); (J.C.H.-G.); (G.A.C.-P.); (J.d.J.E.S.-C.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- American British Cowdray Medical Center, Cancer Center, Mexico City 01120, Mexico
| |
Collapse
|
81
|
Hills KE, Kostarelos K, Wykes RC. Converging Mechanisms of Epileptogenesis and Their Insight in Glioblastoma. Front Mol Neurosci 2022; 15:903115. [PMID: 35832394 PMCID: PMC9271928 DOI: 10.3389/fnmol.2022.903115] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most common and advanced form of primary malignant tumor occurring in the adult central nervous system, and it is frequently associated with epilepsy, a debilitating comorbidity. Seizures are observed both pre- and post-surgical resection, indicating that several pathophysiological mechanisms are shared but also prompting questions about how the process of epileptogenesis evolves throughout GBM progression. Molecular mutations commonly seen in primary GBM, i.e., in PTEN and p53, and their associated downstream effects are known to influence seizure likelihood. Similarly, various intratumoral mechanisms, such as GBM-induced blood-brain barrier breakdown and glioma-immune cell interactions within the tumor microenvironment are also cited as contributing to network hyperexcitability. Substantial alterations to peri-tumoral glutamate and chloride transporter expressions, as well as widespread dysregulation of GABAergic signaling are known to confer increased epileptogenicity and excitotoxicity. The abnormal characteristics of GBM alter neuronal network function to result in metabolically vulnerable and hyperexcitable peri-tumoral tissue, properties the tumor then exploits to favor its own growth even post-resection. It is evident that there is a complex, dynamic interplay between GBM and epilepsy that promotes the progression of both pathologies. This interaction is only more complicated by the concomitant presence of spreading depolarization (SD). The spontaneous, high-frequency nature of GBM-associated epileptiform activity and SD-associated direct current (DC) shifts require technologies capable of recording brain signals over a wide bandwidth, presenting major challenges for comprehensive electrophysiological investigations. This review will initially provide a detailed examination of the underlying mechanisms that promote network hyperexcitability in GBM. We will then discuss how an investigation of these pathologies from a network level, and utilization of novel electrophysiological tools, will yield a more-effective, clinically-relevant understanding of GBM-related epileptogenesis. Further to this, we will evaluate the clinical relevance of current preclinical research and consider how future therapeutic advancements may impact the bidirectional relationship between GBM, SDs, and seizures.
Collapse
Affiliation(s)
- Kate E. Hills
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Catalan Institute for Nanoscience and Nanotechnology (ICN2), Edifici ICN2, Campus UAB, Barcelona, Spain
| | - Robert C. Wykes
- Nanomedicine Lab, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom
- *Correspondence: Robert C. Wykes
| |
Collapse
|
82
|
Alnefaie A, Albogami S, Asiri Y, Ahmad T, Alotaibi SS, Al-Sanea MM, Althobaiti H. Chimeric Antigen Receptor T-Cells: An Overview of Concepts, Applications, Limitations, and Proposed Solutions. Front Bioeng Biotechnol 2022; 10:797440. [PMID: 35814023 PMCID: PMC9256991 DOI: 10.3389/fbioe.2022.797440] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Adaptive immunity, orchestrated by B-cells and T-cells, plays a crucial role in protecting the body from pathogenic invaders and can be used as tools to enhance the body's defense mechanisms against cancer by genetically engineering these immune cells. Several strategies have been identified for cancer treatment and evaluated for their efficacy against other diseases such as autoimmune and infectious diseases. One of the most advanced technologies is chimeric antigen receptor (CAR) T-cell therapy, a pioneering therapy in the oncology field. Successful clinical trials have resulted in the approval of six CAR-T cell products by the Food and Drug Administration for the treatment of hematological malignancies. However, there have been various obstacles that limit the use of CAR T-cell therapy as the first line of defense mechanism against cancer. Various innovative CAR-T cell therapeutic designs have been evaluated in preclinical and clinical trial settings and have demonstrated much potential for development. Such trials testing the suitability of CARs against solid tumors and HIV are showing promising results. In addition, new solutions have been proposed to overcome the limitations of this therapy. This review provides an overview of the current knowledge regarding this novel technology, including CAR T-cell structure, different applications, limitations, and proposed solutions.
Collapse
Affiliation(s)
- Alaa Alnefaie
- Department of Medical Services, King Faisal Medical Complex, Taif, Saudi Arabia
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Yousif Asiri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Saqer S. Alotaibi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Mohammad M. Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Hisham Althobaiti
- Chief of Medical Department, King Faisal Medical Complex (KFMC), Taif, Saudi Arabia
| |
Collapse
|
83
|
B-Cell-Based Immunotherapy: A Promising New Alternative. Vaccines (Basel) 2022; 10:vaccines10060879. [PMID: 35746487 PMCID: PMC9227543 DOI: 10.3390/vaccines10060879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/23/2022] [Accepted: 05/23/2022] [Indexed: 12/04/2022] Open
Abstract
The field of immunotherapy has undergone radical conceptual changes over the last decade. There are various examples of immunotherapy, including the use of monoclonal antibodies, cancer vaccines, tumor-infecting viruses, cytokines, adjuvants, and autologous T cells carrying chimeric antigen receptors (CARs) that can bind cancer-specific antigens known as adoptive immunotherapy. While a lot has been achieved in the field of T-cell immunotherapy, only a fraction of patients (20%) see lasting benefits from this mode of treatment, which is why there is a critical need to turn our attention to other immune cells. B cells have been shown to play both anti- and pro-tumorigenic roles in tumor tissue. In this review, we shed light on the dual nature of B cells in the tumor microenvironment. Furthermore, we discussed the different factors affecting the biology and function of B cells in tumors. In the third section, we described B-cell-based immunotherapies and their clinical applications and challenges. These current studies provide a springboard for carrying out future mechanistic studies to help us unleash the full potential of B cells in immunotherapy.
Collapse
|
84
|
Rong L, Li N, Zhang Z. Emerging therapies for glioblastoma: current state and future directions. J Exp Clin Cancer Res 2022; 41:142. [PMID: 35428347 PMCID: PMC9013078 DOI: 10.1186/s13046-022-02349-7] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/26/2022] [Indexed: 04/15/2023] Open
Abstract
Glioblastoma (GBM) is the most common high-grade primary malignant brain tumor with an extremely poor prognosis. Given the poor survival with currently approved treatments for GBM, new therapeutic strategies are urgently needed. Advances in decades of investment in basic science of glioblastoma are rapidly translated into innovative clinical trials, utilizing improved genetic and epigenetic profiling of glioblastoma as well as the brain microenvironment and immune system interactions. Following these encouraging findings, immunotherapy including immune checkpoint blockade, chimeric antigen receptor T (CAR T) cell therapy, oncolytic virotherapy, and vaccine therapy have offered new hope for improving GBM outcomes; ongoing studies are using combinatorial therapies with the aim of minimizing adverse side-effects and augmenting antitumor immune responses. In addition, techniques to overcome the blood-brain barrier (BBB) for targeted delivery are being tested in clinical trials in patients with recurrent GBM. Here, we set forth the rationales for these promising therapies in treating GBM, review the potential novel agents, the current status of preclinical and clinical trials, and discuss the challenges and future perspectives in glioblastoma immuno-oncology.
Collapse
Affiliation(s)
- Liang Rong
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ni Li
- Institute of Human Virology, Key Laboratory of Tropical Diseases Control Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhenzhen Zhang
- Key Laboratory of Brain, Cognition and Education Science, Ministry of Education, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China.
| |
Collapse
|
85
|
Hussain A. Therapeutic applications of engineered chimeric antigen receptors-T cell for cancer therapy. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2022. [DOI: 10.1186/s43088-022-00238-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Findings of new targeted treatments with adequate safety evaluations are essential for better cancer cures and mortality rates. Immunotherapy holds promise for patients with relapsed disease, with the ability to elicit long-term remissions. Emerging promising clinical results in B-cell malignancy using gene-altered T-lymphocytes uttering chimeric antigen receptors have sparked a lot of interest. This treatment could open the path for a major difference in the way we treat tumors that are resistant or recurring.
Main body
Genetically altered T cells used to produce tumor-specific chimeric antigen receptors are resurrected fields of adoptive cell therapy by demonstrating remarkable success in the treatment of malignant tumors. Because of the molecular complexity of chimeric antigen receptors-T cells, a variety of engineering approaches to improve safety and effectiveness are necessary to realize larger therapeutic uses. In this study, we investigate new strategies for enhancing chimeric antigen receptors-T cell therapy by altering chimeric antigen receptors proteins, T lymphocytes, and their relations with another solid tumor microenvironment (TME) aspects. Furthermore, examine the potential region of chimeric antigen receptors-T cells therapy to become a most effective treatment modality, taking into account the basic and clinical and practical aspect.
Short conclusions
Chimeric antigen receptors-T cells have shown promise in the therapy of hematological cancers. Recent advancements in protein and cell editing, as well as genome-editing technologies, have paved the way for multilayered T cell therapy techniques that can address numerous important demands. At around the same time, there is crosstalk between various intended aspects within the chimeric antigen receptors-T cell diverse biological complexity and possibilities. These breakthroughs substantially improve the ability to comprehend these complex interactions in future solid tumor chimeric antigen receptor-T cell treatment and open up new treatment options for patients that are currently incurable.
Collapse
|
86
|
Otani Y, Yoo JY, Shimizu T, Kurozumi K, Date I, Kaur B. Implications of immune cells in oncolytic herpes simplex virotherapy for glioma. Brain Tumor Pathol 2022; 39:57-64. [PMID: 35384530 DOI: 10.1007/s10014-022-00431-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/27/2022] [Indexed: 12/13/2022]
Abstract
Despite current progress in treatment, glioblastoma (GBM) remains a lethal primary malignant tumor of the central nervous system. Although immunotherapy has recently achieved remarkable survival effectiveness in multiple malignancies, none of the immune checkpoint inhibitors (ICIs) for GBM have shown anti-tumor efficacy in clinical trials. GBM has a characteristic immunosuppressive tumor microenvironment (TME) that results in the failure of ICIs. Oncolytic herpes simplex virotherapy (oHSV) is the most advanced United States Food and Drug Administration-approved virotherapy for advanced metastatic melanoma patients. Recently, another oHSV, Delytact®, was granted conditional approval in Japan against GBM, highlighting it as a promising treatment. Since oncolytic virotherapy can recruit abundant immune cells and modify the immune TME, oncolytic virotherapy for immunologically cold GBM will be an attractive therapeutic option for GBM. However, as these immune cells have roles in both anti-tumor and anti-viral immunity, fine-tuning of the TME using oncolytic virotherapy will be important to maximize the therapeutic efficacy. In this review, we discuss the current knowledge of oHSV, with a focus on the role of immune cells as friend or foe in oncolytic virotherapy.
Collapse
Affiliation(s)
- Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Ji Young Yoo
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| | - Toshihiko Shimizu
- Department of Neurosurgery, Matsuyama Shimin Hospital, 2-6-5 Otemachi, Matsuyama, Ehime, 790-0067, Japan
| | - Kazuhiko Kurozumi
- Department of Neurosurgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, TX, 77030, USA
| |
Collapse
|
87
|
Lechpammer M, Rao R, Shah S, Mirheydari M, Bhattacharya D, Koehler A, Toukam DK, Haworth KJ, Pomeranz Krummel D, Sengupta S. Advances in Immunotherapy for the Treatment of Adult Glioblastoma: Overcoming Chemical and Physical Barriers. Cancers (Basel) 2022; 14:1627. [PMID: 35406398 PMCID: PMC8997081 DOI: 10.3390/cancers14071627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, or glioblastoma multiforme (GBM, WHO Grade IV), is a highly aggressive adult glioma. Despite extensive efforts to improve treatment, the current standard-of-care (SOC) regimen, which consists of maximal resection, radiotherapy, and temozolomide (TMZ), achieves only a 12-15 month survival. The clinical improvements achieved through immunotherapy in several extracranial solid tumors, including non-small-cell lung cancer, melanoma, and non-Hodgkin lymphoma, inspired investigations to pursue various immunotherapeutic interventions in adult glioblastoma patients. Despite some encouraging reports from preclinical and early-stage clinical trials, none of the tested agents have been convincing in Phase III clinical trials. One, but not the only, factor that is accountable for the slow progress is the blood-brain barrier, which prevents most antitumor drugs from reaching the target in appreciable amounts. Herein, we review the current state of immunotherapy in glioblastoma and discuss the significant challenges that prevent advancement. We also provide thoughts on steps that may be taken to remediate these challenges, including the application of ultrasound technologies.
Collapse
Affiliation(s)
- Mirna Lechpammer
- Foundation Medicine, Inc., Cambridge, MA 02141, USA;
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Rohan Rao
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Sanjit Shah
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Mona Mirheydari
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Abigail Koehler
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Donatien Kamdem Toukam
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Kevin J. Haworth
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (M.M.); (K.J.H.)
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (R.R.); (D.B.); (A.K.); (D.K.T.)
| |
Collapse
|
88
|
Burns I, Gwynne WD, Suk Y, Custers S, Chaudhry I, Venugopal C, Singh SK. The Road to CAR T-Cell Therapies for Pediatric CNS Tumors: Obstacles and New Avenues. Front Oncol 2022; 12:815726. [PMID: 35155252 PMCID: PMC8829546 DOI: 10.3389/fonc.2022.815726] [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: 11/15/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Pediatric central nervous system (CNS) tumors are the most common solid tumors diagnosed in children and are the leading cause of pediatric cancer-related death. Those who do survive are faced with the long-term adverse effects of the current standard of care treatments of chemotherapy, radiation, and surgery. There is a pressing need for novel therapeutic strategies to treat pediatric CNS tumors more effectively while reducing toxicity - one of these novel modalities is chimeric antigen receptor (CAR) T-cell therapy. Currently approved for use in several hematological malignancies, there are promising pre-clinical and early clinical data that suggest CAR-T cells could transform the treatment of pediatric CNS tumors. There are, however, several challenges that must be overcome to develop safe and effective CAR T-cell therapies for CNS tumors. Herein, we detail these challenges, focusing on those unique to pediatric patients including antigen selection, tumor immunogenicity and toxicity. We also discuss our perspective on future avenues for CAR T-cell therapies and potential combinatorial treatment approaches.
Collapse
Affiliation(s)
- Ian Burns
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - William D Gwynne
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Yujin Suk
- Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada.,Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Stefan Custers
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Iqra Chaudhry
- Department of Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Sheila K Singh
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
89
|
Han MH, Kim CH. Current Immunotherapeutic Approaches for Malignant Gliomas. Brain Tumor Res Treat 2022; 10:1-11. [PMID: 35118842 PMCID: PMC8819466 DOI: 10.14791/btrt.2022.10.e25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/24/2021] [Accepted: 01/17/2022] [Indexed: 11/20/2022] Open
Abstract
Glioblastoma is the most common malignant central nervous system (CNS) tumor (48.3%), with a median survival of only about 14.6 months. Although the CNS is an immune-privileged site, activated T cells can cross the blood-brain barrier. The recent successes of several immunotherapies for various cancers have drawn interest in immunotherapy for treatment of malignant glioma. There have been extensive attempts to evaluate the efficiency of immunotherapy against malignant glioma. Passive immunotherapy for malignant glioma includes monoclonal antibody-mediated immunotherapy, cytokine-mediated therapy, and adoptive cell transfer, also known as chimeric antigen receptor T cell treatment. On the other hand, active immunotherapy, which stimulates the patient’s adaptive immune system against specific tumor-associated antigens, includes cancer vaccines that are divided into peptide vaccines and cell-based vaccines. In addition, there is immune checkpoint blockade therapy, which increases the efficiency of immunotherapy by reducing the resistance of malignant glioma to immunotherapy. Despite centuries of efforts, immunotherapeutic successes for malignant glioma remain limited. However, many clinical trials of adoptive cell transfer immunotherapy on malignant glioma are ongoing, and the outcomes are eagerly awaited. In addition, although there are still several obstacles, current clinical trials using personalized neoantigen-based dendritic cell vaccines offer new hope to glioblastoma patients. Furthermore, immune checkpoint targeted therapy is expected to decipher the mechanism of immunotherapy resistance in malignant glioma in the near future. More studies are needed to increase the efficacy of immunotherapy in malignant glioma. We hope that immunotherapy will become a new treatment of malignant glioma.
Collapse
Affiliation(s)
- Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Korea
| | - Choong Hyun Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Korea.
| |
Collapse
|
90
|
Using chimeric antigen receptor T-cell therapy to fight glioblastoma multiforme: past, present and future developments. J Neurooncol 2021; 156:81-96. [PMID: 34825292 PMCID: PMC8714623 DOI: 10.1007/s11060-021-03902-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022]
Abstract
Introduction Glioblastoma multiforme (GBM) constitutes one of the deadliest tumors to afflict humans, although it is still considered an orphan disease. Despite testing multiple new and innovative therapies in ongoing clinical trials, the median survival for this type of malignancy is less than two years after initial diagnosis, regardless of therapy. One class of promising new therapies are chimeric antigen receptor T cells or CAR-T which have been shown to be very effective at treating refractory liquid tumors such as B-cell malignancies. However, CAR-T effectivity against solid tumors such as GBM has been limited thus far. Methods A Pubmed, Google Scholar, Directory of Open Access Journals, and Web of Science literature search using the terms chimeric antigen receptor or CAR-T, GBM, solid tumor immunotherapy, immunotherapy, and CAR-T combination was performed for publication dates between January 1987 and November 2021. Results In the current review, we present a comprehensive list of CAR-T cells developed to treat GBM, we describe new possible T-cell engineering strategies against GBM while presenting a short introductory history to the reader regarding the origin(s) of this cutting-edge therapy. We have also compiled a unique list of anti-GBM CAR-Ts with their specific protein sequences and their functions as well as an inventory of clinical trials involving CAR-T and GBM. Conclusions The aim of this review is to introduce the reader to the field of T-cell engineering using CAR-Ts to treat GBM and describe the obstacles that may need to be addressed in order to significantly delay the relentless growth of GBM. Supplementary Information The online version contains supplementary material available at 10.1007/s11060-021-03902-8.
Collapse
|
91
|
Whelan R, Hargaden GC, Knox AJS. Modulating the Blood-Brain Barrier: A Comprehensive Review. Pharmaceutics 2021; 13:1980. [PMID: 34834395 PMCID: PMC8618722 DOI: 10.3390/pharmaceutics13111980] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/23/2022] Open
Abstract
The highly secure blood-brain barrier (BBB) restricts drug access to the brain, limiting the molecular toolkit for treating central nervous system (CNS) diseases to small, lipophilic drugs. Development of a safe and effective BBB modulator would revolutionise the treatment of CNS diseases and future drug development in the area. Naturally, the field has garnered a great deal of attention, leading to a vast and diverse range of BBB modulators. In this review, we summarise and compare the various classes of BBB modulators developed over the last five decades-their recent advancements, advantages and disadvantages, while providing some insight into their future as BBB modulators.
Collapse
Affiliation(s)
- Rory Whelan
- School of Biological and Health Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
- Chemical and Structural Biology, Environmental Sustainability and Health Institute, Technological University Dublin, D07 H6K8 Dublin, Ireland
| | - Grainne C. Hargaden
- School of Chemical and Pharmaceutical Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
| | - Andrew J. S. Knox
- School of Biological and Health Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
- Chemical and Structural Biology, Environmental Sustainability and Health Institute, Technological University Dublin, D07 H6K8 Dublin, Ireland
| |
Collapse
|
92
|
Targeting glioblastoma stem cells: The first step of photodynamic therapy. Photodiagnosis Photodyn Ther 2021; 36:102585. [PMID: 34687963 DOI: 10.1016/j.pdpdt.2021.102585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023]
Abstract
Glioblastoma is one of the most malignant types of brain cancer. Evidence suggests that within gliomas there is a small subpopulation of cells with the capacity for self-renewal, called glioma stem cells. These cells could be responsible for tumorigenesis, chemo and radioresistance, and finally for the recurrence of the tumor. Fluorescence-guided resection have improved the results of treatment against this disease, prolonging the survival of patients by a few months. Also, clinical trials have reported potential improvements in the therapeutic response after photodynamic therapy. Thus far, there are few published works that show the response of glioblastoma stem-like cells to photodynamic therapy. Here, we present a brief review exclusively commenting on the therapeutic approaches to eliminate glioblastoma stem cells and on the research publications about this topic of glioblastoma stem cells in relation to photodynamic therapy. It is our hope that this review will be useful to provide an overview about what is known to date on the topic and to promote the generation of new ideas for the eradication of glioblastoma stem cells by photodynamic treatment.
Collapse
|