1
|
Habibi MA, Mirjani MS, Ahmadvand MH, Delbari P, Arab S, Minaee P, Eazi S, Ahmadpour S. The clinical utility of autologous tumor lysate-loaded dendritic cell vaccination for patients with glioma: A systematic review and meta-analysis. Asia Pac J Clin Oncol 2024; 20:671-680. [PMID: 39244742 DOI: 10.1111/ajco.14110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/03/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Dendritic cell (DC) vaccines show promise for glioma treatment, but optimal use remains uncertain. This meta-analysis examined DC vaccine efficacy and safety for gliomas. METHODS This systematic review and meta-analysis study was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses. From the date of inception to October 23, 2023, electronic databases PubMed, Embase, Web of Science, and Scopus have been thoroughly evaluated. RESULTS A total of 12 studies with 998 patients and a mean age ranging from 40.2 to 56 years were included. Across 12 articles, DC vaccine 6-month overall survival (OS) was 100% [95% confidence interval {95%CI}: 100%-100%]. Respectively, 12-month OS reported 75% [95%CI: 65%-85%] but declined to 32% [95%CI: 20%-43%] for 24-month OS. 6- and 12-month progression-free survival reached 49% [95%CI: 21%-77%] and 19% [95%CI:8%-30%]. Studying radiological outcomes shows that complete response and partial response rates were 13% [95%CI: 17%-42%], and 26% [95%CI: 10%-42%], though stable disease reached 33% [95%CI: 15%-51%], suggesting predominant antineoplastic effects. The progressive disease rate also was 24% [95%CI: 9%-57%]. CONCLUSIONS In gliomas, DC vaccinations show a temporary efficacy; stability is more prevalent than regression. Impacts favor decreased resistance to early disease. Enhancing efficacy remains critical. Early therapy can be enhanced by appropriate supplementary therapy integration.
Collapse
Affiliation(s)
- Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Pouria Delbari
- Student Research Committee, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayan Arab
- School of Medicine, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Poriya Minaee
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - SeyedMohammad Eazi
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Sajjad Ahmadpour
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
2
|
Borges F, Laureano RS, Vanmeerbeek I, Sprooten J, Demeulenaere O, Govaerts J, Kinget L, Saraswat S, Beuselinck B, De Vleeschouwer S, Clement P, De Smet F, Sorg RV, Datsi A, Vigneron N, Naulaerts S, Garg AD. Trial watch: anticancer vaccination with dendritic cells. Oncoimmunology 2024; 13:2412876. [PMID: 39398476 PMCID: PMC11469433 DOI: 10.1080/2162402x.2024.2412876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024] Open
Abstract
Dendritic cells (DCs) are critical players at the intersection of innate and adaptive immunity, making them ideal candidates for anticancer vaccine development. DC-based immunotherapies typically involve isolating patient-derived DCs, pulsing them with tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs), and utilizing maturation cocktails to ensure their effective activation. These matured DCs are then reinfused to elicit tumor-specific T-cell responses. While this approach has demonstrated the ability to generate potent immune responses, its clinical efficacy has been limited due to the immunosuppressive tumor microenvironment. Recent efforts have focused on enhancing the immunogenicity of DC-based vaccines, particularly through combination therapies with T cell-targeting immunotherapies. This Trial Watch summarizes recent advances in DC-based cancer treatments, including the development of new preclinical and clinical strategies, and discusses the future potential of DC-based vaccines in the evolving landscape of immuno-oncology.
Collapse
Affiliation(s)
- Francisca Borges
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Raquel S. Laureano
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Octavie Demeulenaere
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Lisa Kinget
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Saurabh Saraswat
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Benoit Beuselinck
- Department of Medical Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium
- Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Paul Clement
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Leuven Institute for Single-Cell Omics (LISCO), KU Leuven, Leuven, Belgium
- Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Rüdiger V. Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Nathalie Vigneron
- Ludwig Institute for Cancer Research and Cellular Genetics Unit, Université de Louvain, Brussels, Belgium
| | - Stefan Naulaerts
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Abhishek D. Garg
- Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Mao M, Yang W, Zhang X. Current mRNA-based vaccine strategies for glioma treatment. Crit Rev Oncol Hematol 2024; 202:104459. [PMID: 39097247 DOI: 10.1016/j.critrevonc.2024.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/05/2024] Open
Abstract
Gliomas are one of the most aggressive types of brain tumors and are associated with high morbidity and mortality rates. Currently, conventional treatments for gliomas such as surgical resection, radiotherapy, and chemotherapy have limited effectiveness, and new approaches are needed to improve patient outcomes. mRNA-based vaccines represent a promising therapeutic strategy for cancer treatment, including gliomas. Recent advances in immunotherapy using mRNA-based dendritic cell vaccines have shown great potential in preclinical and clinical trials. Dendritic cells are professional antigen-presenting cells that play a crucial role in initiating and regulating immune responses. In this review, we summarize the current progress of mRNA-based vaccines for gliomas, with a focus on recent advances in dendritic cell-based mRNA vaccines. We also discuss the feasibility and safety of mRNA-based clinical applications for gliomas.
Collapse
Affiliation(s)
- Mengqian Mao
- Neuroscience & Metabolism Research, Department of Neurosurgery, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Wanchun Yang
- Neuroscience & Metabolism Research, Department of Neurosurgery, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Xuefeng Zhang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, China.
| |
Collapse
|
4
|
Wang H, Medina R, Ye J, Zhang Y, Chakraborty S, Valenzuela A, Uher O, Hadrava Vanova K, Sun M, Sang X, Park DM, Zenka J, Gilbert MR, Pacak K, Zhuang Z. rWTC-MBTA Vaccine Induces Potent Adaptive Immune Responses Against Glioblastomas via Dynamic Activation of Dendritic Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308280. [PMID: 38298111 PMCID: PMC11005728 DOI: 10.1002/advs.202308280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/19/2023] [Indexed: 02/02/2024]
Abstract
Despite strides in immunotherapy, glioblastoma multiforme (GBM) remains challenging due to low inherent immunogenicity and suppressive tumor microenvironment. Converting "cold" GBMs to "hot" is crucial for immune activation and improved outcomes. This study comprehensively characterized a therapeutic vaccination strategy for preclinical GBM models. The vaccine consists of Mannan-BAM-anchored irradiated whole tumor cells, Toll-like receptor ligands [lipoteichoic acid (LTA), polyinosinic-polycytidylic acid (Poly (I:C)), and resiquimod (R-848)], and anti-CD40 agonistic antibody (rWTC-MBTA). Intracranial GBM models (GL261, SB28 cells) are used to evaluate the vaccine efficacy. A substantial number of vaccinated mice exhibited complete regression of GBM tumors in a T-cell-dependent manner, with no significant toxicity. Long-term tumor-specific immune memory is confirmed upon tumor rechallenge. In the vaccine-draining lymph nodes of the SB28 model, rWTC-MBTA vaccination triggered a major rise in conventional dendritic cell type 1 (cDC1) 12 h post-treatment, followed by an increase in conventional dendritic cell type 2 (cDC2), monocyte-derived dendritic cell (moDC), and plasmacytoid dendritic cell (pDC) on Day 5 and Day 13. Enhanced cytotoxicity of CD4+ and CD8+ T cells in vaccinated mice is verified in co-culture with tumor cells. Analyses of immunosuppressive signals (T-cell exhaustion, myeloid-derived suppressor cells (MDSC), M2 macrophages) in the GBM microenvironment suggest potential combinations with other immunotherapies for enhanced efficacy. In conclusion, the authors findings demonstrate that rWTC-MBTA induces potent and long-term adaptive immune responses against GBM.
Collapse
Affiliation(s)
- Herui Wang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Staff Scientist Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100437 Convent Dr.BethesdaMD20892USA
| | - Rogelio Medina
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Juan Ye
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Yaping Zhang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | | | - Alex Valenzuela
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Ondrej Uher
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Katerina Hadrava Vanova
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Mitchell Sun
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Xueyu Sang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Deric M. Park
- John Theurer Cancer CenterHUMCHackensack Meridian School of Medicine92 2nd StHackensackNJ07601USA
| | - Jan Zenka
- Department of Medical BiologyFaculty of ScienceUniversity of South BohemiaČeské Budějovice37005Czech Republic
| | - Mark R. Gilbert
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of Health9000 Rockville PikeBethesdaMD20892USA
| | - Zhengping Zhuang
- Neuro‐Oncology BranchNational Cancer InstituteNational Institutes of HealthBethesdaMaryland10022USA
- Present address:
Senior Investigator Neuro‐Oncology BranchNational Cancer Institute Center for Cancer ResearchNational Institutes of HealthBuilding 37 Room 100037 Convent DrBethesdaMD20892USA
| |
Collapse
|
5
|
Xie W, Peng Z, Zhou X, Xia Q, Chen M, Zheng X, Sun H, Zou H, Xu L, Du Z, Li E, Wu B. The Expression Pattern and Clinical Significance of Lysyl Oxidase Family in Gliomas. DOKL BIOCHEM BIOPHYS 2023; 510:132-143. [PMID: 37582875 DOI: 10.1134/s1607672922600269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 08/17/2023]
Abstract
LOX (Lysyl oxidase) family participates in the catalysis of collagen and elastin to maintain ECM homeostasis. Glioma is the most common primary brain tumor and LOX family has not been systemic studied in glioma. In this study, we found LOX family members are upregulated expressed in gliomas samples. A protein-protein interaction network (PPIN) was construct to visualize and understand the differential expression pattern, as well as functional annotation, for LOX family and their interacting proteins, which involved in collagen fibril organization and MAPK signaling pathway. Through subcellular localization distribution, the LOX family members distribute both intracellular and extracellular. All five LOX members are consistently significantly correlate with dendritic cell both in immune infiltrate of GBM and LGG. Survival analysis showed that high expression of LOX family is associated with a poor prognosis of gliomas patients. These analyses provide important clues to identify the potential biological roles for LOX family in gliomas, which might serve as diagnosis markers.
Collapse
Affiliation(s)
- Weijie Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Zhongte Peng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Xiao Zhou
- Department of Central Laboratory, Shantou Central Hospital, 515041, Shantou, China
| | - Qiaoxi Xia
- Department of Central Laboratory, Shantou Central Hospital, 515041, Shantou, China
| | - Mantong Chen
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Xiaoqi Zheng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Hong Sun
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Haiying Zou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, 515041, Shantou, China
| | - Zepeng Du
- Department of Central Laboratory, Shantou Central Hospital, 515041, Shantou, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China.
| | - Bingli Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, 515041, Shantou, China.
| |
Collapse
|
6
|
Shah S, Famta P, Tiwari V, Kotha AK, Kashikar R, Chougule MB, Chung YH, Steinmetz NF, Uddin M, Singh SB, Srivastava S. Instigation of the epoch of nanovaccines in cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1870. [PMID: 36410742 PMCID: PMC10182210 DOI: 10.1002/wnan.1870] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/03/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022]
Abstract
Cancer is an unprecedented proliferation of cells leading to abnormalities in differentiation and maturation. Treatment of primary and metastatic cancer is challenging. In addition to surgery, chemotherapy and radiation therapies have been conventionally used; however, they suffer from severe toxicity and non-specificity. Immunotherapy, the science of programming the body's own defense system against cancer has gained tremendous attention in the last few decades. However, partial immunogenic stimulation, premature degradation and inability to activate dendritic and helper T cells has resulted in limited clinical success. The era of nanomedicine has brought about several breakthroughs in various pharmaceutical and biomedical fields. Hereby, we review and discuss the interplay of tumor microenvironment (TME) and the immunological cascade and how they can be employed to develop nanoparticle-based cancer vaccines and immunotherapies. Nanoparticles composed of lipids, polymers and inorganic materials contain useful properties suitable for vaccine development. Proteinaceous vaccines derived from mammalian viruses, bacteriophages and plant viruses also have unique advantages due to their immunomodulation capabilities. This review accounts for all such considerations. Additionally, we explore how attributes of nanotechnology can be utilized to develop successful nanomedicine-based vaccines for cancer therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Saurabh Shah
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| | - Paras Famta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering, & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, INDIA
| | - Arun K Kotha
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Rama Kashikar
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Mahavir Bhupal Chougule
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Young Hun Chung
- Departments of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicole F. Steinmetz
- Departments of Bioengineering, NanoEngineering, Radiology, Moores Cancer Center, Center for Nano-ImmunoEngineering, Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mohammad Uddin
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, INDIA
| |
Collapse
|
7
|
Aggarwal P, Luo W, Pehlivan KC, Hoang H, Rajappa P, Cripe TP, Cassady KA, Lee DA, Cairo MS. Pediatric versus adult high grade glioma: Immunotherapeutic and genomic considerations. Front Immunol 2022; 13:1038096. [PMID: 36483545 PMCID: PMC9722734 DOI: 10.3389/fimmu.2022.1038096] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022] Open
Abstract
High grade gliomas are identified as malignant central nervous tumors that spread rapidly and have a universally poor prognosis. Historically high grade gliomas in the pediatric population have been treated similarly to adult high grade gliomas. For the first time, the most recent classification of central nervous system tumors by World Health Organization has divided adult from pediatric type diffuse high grade gliomas, underscoring the biologic differences between these tumors in different age groups. The objective of our review is to compare high grade gliomas in the adult versus pediatric patient populations, highlighting similarities and differences in epidemiology, etiology, pathogenesis and therapeutic approaches. High grade gliomas in adults versus children have varying clinical presentations, molecular biology background, and response to chemotherapy, as well as unique molecular targets. However, increasing evidence show that they both respond to recently developed immunotherapies. This review summarizes the distinctions and commonalities between the two in disease pathogenesis and response to therapeutic interventions with a focus on immunotherapy.
Collapse
Affiliation(s)
- Payal Aggarwal
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | | | - Hai Hoang
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Prajwal Rajappa
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Timothy P. Cripe
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Kevin A. Cassady
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Dean A. Lee
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States,Department of Medicine, New York Medical College, Valhalla, NY, United States,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States,*Correspondence: Mitchell S. Cairo,
| |
Collapse
|
8
|
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
|
9
|
Li L, Zhou J, Dong X, Liao Q, Zhou D, Zhou Y. Dendritic cell vaccines for glioblastoma fail to complete clinical translation: Bottlenecks and potential countermeasures. Int Immunopharmacol 2022; 109:108929. [PMID: 35700581 DOI: 10.1016/j.intimp.2022.108929] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/20/2022] [Accepted: 06/05/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma (GBM) is a heterogeneous and invasive WHO grade IV brain tumor. Patients with GBM have a median overall survival (OS) of only 14 to 17 months when treated with surgical resection and chemoradiation. As one of the most promising anti-tumor immunotherapies, dendritic cell (DC) vaccines have demonstrated good efficacy, safety, and tolerability in many clinical trials. However, to date, no Phase III clinical trial has achieved positive endpoints and truly implement clinical development and transformation. Moreover, the survival benefits of DC vaccines for patients with GBM seem to have a delayed effect; therefore, we urgently require strategies to optimize DC vaccines to advance the time point of its survival benefits. Here, we discuss the latest clinical trial progress of DC vaccines in GBM and summarize the benefits and drawbacks of various vaccine design options, as well as the challenges faced in clinical translation. Moreover, we target future combination therapy strategies for DC vaccines in GBM, which provides a new perspective for comprehensively understanding the effectiveness, limitations, and new directions of the development of DC vaccines.
Collapse
Affiliation(s)
- Luohong Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Jing Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Xueting Dong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Dongbo Zhou
- Department of Geriatric, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China, Hunan 410008, China.
| | - Yanhong Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China; Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, Hunan 410078, China; Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
10
|
Laureano RS, Sprooten J, Vanmeerbeerk I, Borras DM, Govaerts J, Naulaerts S, Berneman ZN, Beuselinck B, Bol KF, Borst J, Coosemans A, Datsi A, Fučíková J, Kinget L, Neyns B, Schreibelt G, Smits E, Sorg RV, Spisek R, Thielemans K, Tuyaerts S, De Vleeschouwer S, de Vries IJM, Xiao Y, Garg AD. Trial watch: Dendritic cell (DC)-based immunotherapy for cancer. Oncoimmunology 2022; 11:2096363. [PMID: 35800158 PMCID: PMC9255073 DOI: 10.1080/2162402x.2022.2096363] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/01/2022] [Accepted: 06/28/2022] [Indexed: 12/21/2022] Open
Abstract
Dendritic cell (DC)-based vaccination for cancer treatment has seen considerable development over recent decades. However, this field is currently in a state of flux toward niche-applications, owing to recent paradigm-shifts in immuno-oncology mobilized by T cell-targeting immunotherapies. DC vaccines are typically generated using autologous (patient-derived) DCs exposed to tumor-associated or -specific antigens (TAAs or TSAs), in the presence of immunostimulatory molecules to induce DC maturation, followed by reinfusion into patients. Accordingly, DC vaccines can induce TAA/TSA-specific CD8+/CD4+ T cell responses. Yet, DC vaccination still shows suboptimal anti-tumor efficacy in the clinic. Extensive efforts are ongoing to improve the immunogenicity and efficacy of DC vaccines, often by employing combinatorial chemo-immunotherapy regimens. In this Trial Watch, we summarize the recent preclinical and clinical developments in this field and discuss the ongoing trends and future perspectives of DC-based immunotherapy for oncological indications.
Collapse
Affiliation(s)
- Raquel S Laureano
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jenny Sprooten
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeerk
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Daniel M Borras
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jannes Govaerts
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Zwi N Berneman
- Department of Haematology, Antwerp University Hospital, Edegem, Belgium
- Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | | | - Kalijn F Bol
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jannie Borst
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - an Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, Ku Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Jitka Fučíková
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Lisa Kinget
- Department of General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - Bart Neyns
- Department of Medical Oncology, UZ Brussel, Brussels, Belgium
| | - Gerty Schreibelt
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, The Netherlands
| | - Evelien Smits
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
- Center for Oncological Research, Integrated Personalized and Precision Oncology Network, University of Antwerp, Wilrijk, Belgium
| | - Rüdiger V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, University Hospital Motol, Prague, Czech Republic
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sandra Tuyaerts
- Department of Medical Oncology, UZ Brussel, Brussels, Belgium
- Laboratory of Medical and Molecular Oncology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - I Jolanda M de Vries
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences; Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yanling Xiao
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Challenges in glioblastoma immunotherapy: mechanisms of resistance and therapeutic approaches to overcome them. Br J Cancer 2022; 127:976-987. [DOI: 10.1038/s41416-022-01864-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/23/2022] [Accepted: 05/12/2022] [Indexed: 11/08/2022] Open
|
12
|
Malik DG, Rath TJ, Urcuyo Acevedo JC, Canoll PD, Swanson KR, Boxerman JL, Quarles CC, Schmainda KM, Burns TC, Hu LS. Advanced MRI Protocols to Discriminate Glioma From Treatment Effects: State of the Art and Future Directions. FRONTIERS IN RADIOLOGY 2022; 2:809373. [PMID: 37492687 PMCID: PMC10365126 DOI: 10.3389/fradi.2022.809373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/01/2022] [Indexed: 07/27/2023]
Abstract
In the follow-up treatment of high-grade gliomas (HGGs), differentiating true tumor progression from treatment-related effects, such as pseudoprogression and radiation necrosis, presents an ongoing clinical challenge. Conventional MRI with and without intravenous contrast serves as the clinical benchmark for the posttreatment surveillance imaging of HGG. However, many advanced imaging techniques have shown promise in helping better delineate the findings in indeterminate scenarios, as posttreatment effects can often mimic true tumor progression on conventional imaging. These challenges are further confounded by the histologic admixture that can commonly occur between tumor growth and treatment-related effects within the posttreatment bed. This review discusses the current practices in the surveillance imaging of HGG and the role of advanced imaging techniques, including perfusion MRI and metabolic MRI.
Collapse
Affiliation(s)
- Dania G. Malik
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - Tanya J. Rath
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
| | - Javier C. Urcuyo Acevedo
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| | - Peter D. Canoll
- Departments of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Kristin R. Swanson
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| | - Jerrold L. Boxerman
- Department of Diagnostic Imaging, Brown University, Providence, RI, United States
| | - C. Chad Quarles
- Department of Neuroimaging Research & Barrow Neuroimaging Innovation Center, Barrow Neurologic Institute, Phoenix, AZ, United States
| | - Kathleen M. Schmainda
- Department of Biophysics & Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Terry C. Burns
- Departments of Neurologic Surgery and Neuroscience, Mayo Clinic, Rochester, MN, United States
| | - Leland S. Hu
- Department of Radiology, Mayo Clinic, Phoenix, AZ, United States
- Mathematical Neurooncology Lab, Precision Neurotherapeutics Innovation Program, Mayo Clinic, Phoenix, AZ, United States
| |
Collapse
|
13
|
Codrici E, Popescu ID, Tanase C, Enciu AM. Friends with Benefits: Chemokines, Glioblastoma-Associated Microglia/Macrophages, and Tumor Microenvironment. Int J Mol Sci 2022; 23:ijms23052509. [PMID: 35269652 PMCID: PMC8910233 DOI: 10.3390/ijms23052509] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022] Open
Abstract
Glioma is the most common primary intracranial tumor and has the greatest prevalence of all brain tumors. Treatment resistance and tumor recurrence in GBM are mostly explained by considerable alterations within the tumor microenvironment, as well as extraordinary cellular and molecular heterogeneity. Soluble factors, extracellular matrix components, tissue-resident cell types, resident or newly recruited immune cells together make up the GBM microenvironment. Regardless of many immune cells, a profound state of tumor immunosuppression is supported and developed, posing a considerable hurdle to cancer cells' immune-mediated destruction. Several studies have suggested that various GBM subtypes present different modifications in their microenvironment, although the importance of the microenvironment in treatment response has yet to be determined. Understanding the microenvironment and how it changes after therapies is critical because it can influence the remaining invasive GSCs and lead to recurrence. This review article sheds light on the various components of the GBM microenvironment and their roles in tumoral development, as well as immune-related biological processes that support the interconnection/interrelationship between different cell types. Also, we summarize the current understanding of the modulation of soluble factors and highlight the dysregulated inflammatory chemokine/specific receptors cascades/networks and their significance in tumorigenesis, cancer-related inflammation, and metastasis.
Collapse
Affiliation(s)
- Elena Codrici
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Correspondence: (E.C.); (I.-D.P.); (A.-M.E.)
| | - Ionela-Daniela Popescu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Correspondence: (E.C.); (I.-D.P.); (A.-M.E.)
| | - Cristiana Tanase
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Department of Clinical Biochemistry, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Ana-Maria Enciu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Correspondence: (E.C.); (I.-D.P.); (A.-M.E.)
| |
Collapse
|
14
|
Delayed Effect of Dendritic Cells Vaccination on Survival in Glioblastoma: A Systematic Review and Meta-Analysis. Curr Oncol 2022; 29:881-891. [PMID: 35200574 PMCID: PMC8870360 DOI: 10.3390/curroncol29020075] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Dendritic cell vaccination (DCV) strategies, thanks to a complex immune response, may flare tumor regression and improve patients’ long-term survival. This meta-analysis aims to assess the efficacy of DCV for newly diagnosed glioblastoma patients in clinical trials. Methods: The study databases, including PubMed, Web of Knowledge, Google Scholar, Scopus, and Cochrane, were searched by two blinded investigators considering eligible studies based on the following keywords: “glioblastoma multiforme”, “dendritic cell”, “vaccination”, “immunotherapy”, “immune system”, “immune response”, “chemotherapy”, “recurrence”, and “temozolomide”. Among the 157 screened, only 15 articles were eligible for the final analysis. Results: Regimens including DCV showed no effect on 6-month progression-free survival (PFS, HR = 1.385, 95% CI: 0.822–2.335, p = 0.673) or on 6-month overall survival (OS, HR = 1.408, 95% CI: 0.882–2.248, p = 0.754). In contrast, DCV led to significantly longer 1-year OS (HR = 1.936, 95% CI: 1.396–2.85, p = 0.001) and longer 2-year OS (HR = 3.670, 95% CI: 2.291–5.879, p = 0.001) versus control groups. Hence, introducing DCV could lead to increased 1 and 2-year survival of patients by 1.9 and 3.6 times, respectively. Conclusion: Antitumor regimens including DCV can effectively improve mid-term survival in patients suffering glioblastoma multiforme (GBM), but its impact emerges only after one year from vaccination. These data indicate the need for more time to achieve an anti-GBM immune response and suggest additional therapeutics, such as checkpoint inhibitors, to empower an earlier DCV action in patients affected by a very poor prognosis.
Collapse
|
15
|
Gonçalves FG, Viaene AN, Vossough A. Advanced Magnetic Resonance Imaging in Pediatric Glioblastomas. Front Neurol 2021; 12:733323. [PMID: 34858308 PMCID: PMC8631300 DOI: 10.3389/fneur.2021.733323] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/12/2021] [Indexed: 12/26/2022] Open
Abstract
The shortly upcoming 5th edition of the World Health Organization Classification of Tumors of the Central Nervous System is bringing extensive changes in the terminology of diffuse high-grade gliomas (DHGGs). Previously "glioblastoma," as a descriptive entity, could have been applied to classify some tumors from the family of pediatric or adult DHGGs. However, now the term "glioblastoma" has been divested and is no longer applied to tumors in the family of pediatric types of DHGGs. As an entity, glioblastoma remains, however, in the family of adult types of diffuse gliomas under the insignia of "glioblastoma, IDH-wildtype." Of note, glioblastomas still can be detected in children when glioblastoma, IDH-wildtype is found in this population, despite being much more common in adults. Despite the separation from the family of pediatric types of DHGGs, what was previously labeled as "pediatric glioblastomas" still remains with novel labels and as new entities. As a result of advances in molecular biology, most of the previously called "pediatric glioblastomas" are now classified in one of the four family members of pediatric types of DHGGs. In this review, the term glioblastoma is still apocryphally employed mainly due to its historical relevance and the paucity of recent literature dealing with the recently described new entities. Therefore, "glioblastoma" is used here as an umbrella term in the attempt to encompass multiple entities such as astrocytoma, IDH-mutant (grade 4); glioblastoma, IDH-wildtype; diffuse hemispheric glioma, H3 G34-mutant; diffuse pediatric-type high-grade glioma, H3-wildtype and IDH-wildtype; and high grade infant-type hemispheric glioma. Glioblastomas are highly aggressive neoplasms. They may arise anywhere in the developing central nervous system, including the spinal cord. Signs and symptoms are non-specific, typically of short duration, and usually derived from increased intracranial pressure or seizure. Localized symptoms may also occur. The standard of care of "pediatric glioblastomas" is not well-established, typically composed of surgery with maximal safe tumor resection. Subsequent chemoradiation is recommended if the patient is older than 3 years. If younger than 3 years, surgery is followed by chemotherapy. In general, "pediatric glioblastomas" also have a poor prognosis despite surgery and adjuvant therapy. Magnetic resonance imaging (MRI) is the imaging modality of choice for the evaluation of glioblastomas. In addition to the typical conventional MRI features, i.e., highly heterogeneous invasive masses with indistinct borders, mass effect on surrounding structures, and a variable degree of enhancement, the lesions may show restricted diffusion in the solid components, hemorrhage, and increased perfusion, reflecting increased vascularity and angiogenesis. In addition, magnetic resonance spectroscopy has proven helpful in pre- and postsurgical evaluation. Lastly, we will refer to new MRI techniques, which have already been applied in evaluating adult glioblastomas, with promising results, yet not widely utilized in children.
Collapse
Affiliation(s)
- Fabrício Guimarães Gonçalves
- Division of Neuroradiology, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Arastoo Vossough
- Division of Neuroradiology, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, United States.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
16
|
Rasheed S, Rehman K, Akash MSH. An insight into the risk factors of brain tumors and their therapeutic interventions. Biomed Pharmacother 2021; 143:112119. [PMID: 34474351 DOI: 10.1016/j.biopha.2021.112119] [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: 07/02/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022] Open
Abstract
Brain tumors are an abnormal growth of cells in the brain, also known as multifactorial groups of neoplasm. Incidence rates of brain tumors increase rapidly, and it has become a leading cause of tumor related deaths globally. Several factors have potential risks for intracranial neoplasm. To date, the International Agency for Research on Cancer has classified the ionizing radiation and the N-nitroso compounds as established carcinogens and probable carcinogens respectively. Diagnosis of brain tumors is based on histopathology and suitable imaging techniques. Labeled amino acids and fluorodeoxyglucose with or without contrast-enhanced MRI are used for the evaluation of tumor traces. T2-weighted MRI is an advanced diagnostic implementation, used for the detection of low-grade gliomas. Treatment decisions are based on tumor size, location, type, patient's age and health status. Conventional therapeutic approaches for tumor treatment are surgery, radiotherapy and chemotherapy. While the novel strategies may include targeted therapy, electric field treatments and vaccine therapy. Inhibition of cyclin-dependent kinase inhibitors is an attractive tumor mitigation strategy for advanced-stage cancers; in the future, it may prove to be a useful targeted therapy. The blood-brain barrier poses a major hurdle in the transport of therapeutics towards brain tissues. Moreover, nanomedicine has gained a vital role in cancer therapy. Nano drug delivery system such as liposomal drug delivery has been widely used in the cancer treatment. Liposome encapsulated drugs have improved therapeutic efficacy than free drugs. Numerous treatment therapies for brain tumors are in advanced clinical research.
Collapse
Affiliation(s)
- Sumbal Rasheed
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
17
|
Schaller TH, Snyder DJ, Spasojevic I, Gedeon PC, Sanchez-Perez L, Sampson JH. First in human dose calculation of a single-chain bispecific antibody targeting glioma using the MABEL approach. J Immunother Cancer 2021; 8:jitc-2019-000213. [PMID: 32273346 PMCID: PMC7254109 DOI: 10.1136/jitc-2019-000213] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background First-in-human (FIH) clinical trials require careful selection of a safe yet biologically relevant starting dose. Typically, such starting doses are selected based on toxicity studies in a pharmacologically relevant animal model. However, with the advent of target-specific and highly active immunotherapeutics, both the Food and Drug Administration and the European Medicines Agency have provided guidance that recommend determining a safe starting dose based on a minimum anticipated biological effect level (MABEL) approach. Methods We recently developed a T cell activating bispecific antibody that effectively treats orthotopic patient-derived malignant glioma and syngeneic glioblastoma in mice (hEGFRvIII:CD3 bi-scFv). hEGFRvIII:CD3 bi-scFv is comprized of two single chain antibody fragments (bi-scFvs) that bind mutant epidermal growth factor receptor variant III (EGFRvIII), a mutation frequently seen in malignant glioma, and human CD3ε on T cells, respectively. In order to establish a FIH dose, we used a MABEL approach to select a safe starting dose for hEGFRvIII:CD3 bi-scFv, based on a combination of in vitro data, in vivo animal studies, and theoretical human receptor occupancy modeling. Results Using the most conservative approach to the MABEL assessment, a dose of 57.4 ng hEGFRvIII:CD3 bi-scFv/kg body weight was selected as a safe starting dose for a FIH clinical study. Conclusions The comparison of our MABEL-based starting dose to our in vivo efficacious dose and the theoretical human receptor occupancy strongly supports that our human starting dose of 57.4 ng hEGFRvIII:CD3 bi-scFv/patient kg will be safe.
Collapse
Affiliation(s)
- Teilo H Schaller
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States.,Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States
| | - David J Snyder
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States
| | - Ivan Spasojevic
- PK/PD Core Laboratory, Duke Cancer Institute, Durham, North Carolina, United States.,Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States
| | - Patrick C Gedeon
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States
| | - Luis Sanchez-Perez
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States.,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States
| | - John H Sampson
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, United States .,Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, United States.,Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
18
|
Han J, Sun J, Zhang G, Chen H. DCs-based therapies: potential strategies in severe SARS-CoV-2 infection. Int J Med Sci 2021; 18:406-418. [PMID: 33390810 PMCID: PMC7757148 DOI: 10.7150/ijms.47706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/09/2020] [Indexed: 01/08/2023] Open
Abstract
Pneumonia caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is spreading globally. There have been strenuous efforts to reveal the mechanisms that the host defends itself against invasion by this virus. The immune system could play a crucial role in virus infection. Dendritic cell as sentinel of the immune system plays an irreplaceable role. Dendritic cells-based therapeutic approach may be a potential strategy for SARS-CoV-2 infection. In this review, the characteristics of coronavirus are described briefly. We focus on the essential functions of dendritic cell in severe SARS-CoV-2 infection. Basis of treatment based dendritic cells to combat coronavirus infections is summarized. Finally, we propose that the combination of DCs based vaccine and other therapy is worth further study.
Collapse
Affiliation(s)
- Jian Han
- General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine of Dalian Medical University, Dalian 116044, China
- Department of Pharmaceutical Sciences USF Health, Taneja College of Pharmacy University of South Florida, Tampa, FL, USA
| | - Jiazhi Sun
- Department of Pharmaceutical Sciences USF Health, Taneja College of Pharmacy University of South Florida, Tampa, FL, USA
| | - Guixin Zhang
- General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine of Dalian Medical University, Dalian 116044, China
| | - Hailong Chen
- General Surgery Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute of Integrative Medicine of Dalian Medical University, Dalian 116044, China
| |
Collapse
|
19
|
Garcia-Fabiani MB, Ventosa M, Comba A, Candolfi M, Nicola Candia AJ, Alghamri MS, Kadiyala P, Carney S, Faisal SM, Schwendeman A, Moon JJ, Scheetz L, Lahann J, Mauser A, Lowenstein PR, Castro MG. Immunotherapy for gliomas: shedding light on progress in preclinical and clinical development. Expert Opin Investig Drugs 2020; 29:659-684. [PMID: 32400216 DOI: 10.1080/13543784.2020.1768528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Gliomas are infiltrating brain tumors associated with high morbidity and mortality. Current standard of care includes radiation, chemotherapy, and surgical resection. Today, survival rates for malignant glioma patients remain dismal and unchanged for decades. The glioma microenvironment is highly immunosuppressive and consequently this has motivated the development of immunotherapies for counteracting this condition, enabling the immune cells within the tumor microenvironment to react against this tumor. AREAS COVERED The authors discuss immunotherapeutic strategies for glioma in phase-I/II clinical trials and illuminate their mechanisms of action, limitations, and key challenges. They also examine promising approaches under preclinical development. EXPERT OPINION In the last decade there has been an expansion in immune-mediated anti-cancer therapies. In the glioma field, sophisticated strategies have been successfully implemented in preclinical models. Unfortunately, clinical trials have not yet yielded consistent results for glioma patients. This could be attributed to our limited understanding of the complex immune cell infiltration and its interaction with the tumor cells, the selected time for treatment, the combination with other therapies and the route of administration of the agent. Applying these modalities to treat malignant glioma is challenging, but many new alternatives are emerging to by-pass these hurdles.
Collapse
Affiliation(s)
- Maria B Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Maria Ventosa
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Alejandro J Nicola Candia
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Cancer Biology Graduate Program, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - Joerg Lahann
- Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Chemical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Ava Mauser
- Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Chemical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
20
|
Jeanmougin M, Håvik AB, Cekaite L, Brandal P, Sveen A, Meling TR, Ågesen TH, Scheie D, Heim S, Lothe RA, Lind GE. Improved prognostication of glioblastoma beyond molecular subtyping by transcriptional profiling of the tumor microenvironment. Mol Oncol 2020; 14:1016-1027. [PMID: 32171051 PMCID: PMC7191188 DOI: 10.1002/1878-0261.12668] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 01/17/2020] [Accepted: 03/12/2020] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma (GBM), the most aggressive form of brain cancer, is characterized by a high level of molecular heterogeneity, and infiltration by various immune and stromal cell populations. Important advances have been made in deciphering the microenvironment of GBMs, but its association with existing molecular subtypes and its potential prognostic role remain elusive. We have investigated the abundance of infiltrating immune and stromal cells in silico, from gene expression profiles. Two cohorts, including in‐house normal brain and glioma samples (n = 70) and a large sample set from TCGA (n = 393), were combined into a single exploratory dataset. A third independent cohort (n = 124) was used for validation. Tumors were clustered based on their microenvironment infiltration profiles, and associations with known GBM molecular subtypes and patient outcome were tested a posteriori in a multivariable setting. We identified a subset of GBM samples with significantly higher abundances of most immune and stromal cell populations. This subset showed increased expression of both immune suppressor and immune effector genes compared to other GBMs and was enriched for the mesenchymal molecular subtype. Survival analyses suggested that tumor microenvironment infiltration pattern was an independent prognostic factor for GBM patients. Among all, patients with the mesenchymal subtype with low immune and stromal infiltration had the poorest survival. By combining molecular subtyping with gene expression measures of tumor infiltration, the present work contributes with improving prognostic models in GBM.
Collapse
Affiliation(s)
- Marine Jeanmougin
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - Annette B Håvik
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway.,Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - Lina Cekaite
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - Petter Brandal
- Department of Oncology, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Torstein R Meling
- Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Trude H Ågesen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - David Scheie
- Department of Pathology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Sverre Heim
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Guro E Lind
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
21
|
Wei BM, Hanlon D, Khalil D, Han P, Tatsuno K, Sobolev O, Edelson RL. Extracorporeal Photochemotherapy: Mechanistic Insights Driving Recent Advances and Future Directions. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:145-159. [PMID: 32226344 PMCID: PMC7087063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells, necessary for the initiation and maintenance of antigen-specific immunity and tolerance. Decades of research have been driven by hopes to harness the immunological capabilities of DCs and achieve physiological partnership with the immune system for therapeutic ends. Potential applications for DC-based immunotherapy include treatments for cancer, autoimmune disorders, and infectious diseases. However, DCs have poor availability in peripheral and lymphoid tissues and have poor survivability in culture, leading to the development of multiple strategies to generate and manipulate large numbers of DCs ex vivo. Among these is Extracorporeal Photopheresis (ECP), a widely used cancer immunotherapy. Recent advancements have uncovered that stimulation of monocyte-to-DC maturation via physiologic inflammatory signaling lies at the mechanistic core of ECP. Here, we describe the landscape of DC-based immunotherapy, the historical context of ECP, the current mechanistic understanding of ex vivo monocyte-to-DC maturation in ECP, and the implications of this understanding on making scientifically driven improvements to modern ECP protocols and devices.
Collapse
Affiliation(s)
- Brian M. Wei
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Douglas Hanlon
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - David Khalil
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Patrick Han
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT
| | - Kazuki Tatsuno
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Olga Sobolev
- Department of Dermatology, Yale School of Medicine, New Haven, CT
| | - Richard L. Edelson
- Department of Dermatology, Yale School of Medicine, New Haven, CT,To whom all correspondence should be addressed: Richard L. Edelson, MD, PO Box 208059, 333 Cedar St., New Haven, CT, 06520-8059; Tel: 203-785-4092, Fax: 203-737-5318,
| |
Collapse
|
22
|
Xu S, Tang L, Li X, Fan F, Liu Z. Immunotherapy for glioma: Current management and future application. Cancer Lett 2020; 476:1-12. [PMID: 32044356 DOI: 10.1016/j.canlet.2020.02.002] [Citation(s) in RCA: 435] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/30/2022]
Abstract
Gliomas are intrinsic brain tumors that originate from neuroglial progenitor cells. Conventional therapies, including surgery, chemotherapy, and radiotherapy, have achieved limited improvements in the prognosis of glioma patients. Immunotherapy, a revolution in cancer treatment, has become a promising strategy with the ability to penetrate the blood-brain barrier since the pioneering discovery of lymphatics in the central nervous system. Here we detail the current management of gliomas and previous studies assessing different immunotherapies in gliomas, despite the fact that the associated clinical trials have not been completed yet. Moreover, several drugs that have undergone clinical trials are listed as novel strategies for future application; however, these clinical trials have indicated limited efficacy in glioma. Therefore, additional studies are warranted to evaluate novel therapeutic approaches in glioma treatment.
Collapse
Affiliation(s)
- Shengchao Xu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Lu Tang
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xizhe Li
- Department of Thoracic Surgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Fan Fan
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
23
|
Guerra-García P, Marshall LV, Cockle JV, Ramachandran PV, Saran FH, Jones C, Carceller F. Challenging the indiscriminate use of temozolomide in pediatric high-grade gliomas: A review of past, current, and emerging therapies. Pediatr Blood Cancer 2020; 67:e28011. [PMID: 31617673 DOI: 10.1002/pbc.28011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/31/2019] [Accepted: 09/11/2019] [Indexed: 01/26/2023]
Abstract
Pediatric high-grade gliomas (pHGG) constitute 8% to 12% of primary brain tumors in childhood. The most widely utilized treatment encompasses surgical resection followed by focal radiotherapy and temozolomide. However, experiences over past decades have not demonstrated improved outcomes. pHGG have been classified into different molecular subgroups defined by mutations in histone 3, IDH gene, MAPK pathway, and others, thereby providing a rationale for various targeted therapies. Additionally, immunotherapy and drug repurposing have also become attractive adjunctive treatments. This review focuses on past, present, and emerging treatments for pHGG integrating molecular research with the mainstream pediatric drug development in Europe and the United States to sketch a way forward in the development of novel therapeutic approaches. The implementation of randomized clinical trials with adaptive designs, underpinned by a robust biological rationale, and harnessing collaboration between the pharmaceutical industry, academia, regulators and patients/parents organizations will be essential to improve the outcomes for these children.
Collapse
Affiliation(s)
- Pilar Guerra-García
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.,Paediatric Oncology Department, 12 de Octubre University Hospital, Madrid, Spain
| | - Lynley V Marshall
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.,Division of Clinical Studies, Institute of Cancer Research, London, United Kingdom
| | - Julia V Cockle
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | | | - Frank H Saran
- Department of Radiation Oncology, The Royal Marsden NHS Foundation Trust, London, United Kingdom.,Department of Radiation Oncology, Auckland District Health Board, Auckland, New Zealand
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, London, United Kingdom
| | - Fernando Carceller
- Children and Young People's Unit, The Royal Marsden NHS Foundation Trust, Sutton, United Kingdom.,Division of Clinical Studies, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
24
|
Han P, Hanlon D, Sobolev O, Chaudhury R, Edelson RL. Ex vivo dendritic cell generation-A critical comparison of current approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:251-307. [PMID: 31759433 DOI: 10.1016/bs.ircmb.2019.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells, required for the initiation of naïve and memory T cell responses and regulation of adaptive immunity. The discovery of DCs in 1973, which culminated in the Nobel Prize in Physiology or Medicine in 2011 for Ralph Steinman and colleagues, initially focused on the identification of adherent mononuclear cell fractions with uniquely stellate dendritic morphology, followed by key discoveries of their critical immunologic role in initiating and maintaining antigen-specific immunity and tolerance. The medical promise of marshaling these key capabilities of DCs for therapeutic modulation of antigen-specific immune responses has guided decades of research in hopes to achieve genuine physiologic partnership with the immune system. The potential uses of DCs in immunotherapeutic applications include cancer, infectious diseases, and autoimmune disorders; thus, methods for rapid and reliable large-scale production of DCs have been of great academic and clinical interest. However, difficulties in obtaining DCs from lymphoid and peripheral tissues, low numbers and poor survival in culture, have led to advancements in ex vivo production of DCs, both for probing molecular details of DC function as well as for experimenting with their clinical utility. Here, we review the development of a diverse array of DC production methodologies, ranging from cytokine-based strategies to genetic engineering tools devised for enhancing DC-specific immunologic functions. Further, we explore the current state of DC therapies in clinic, as well as emerging insights into physiologic production of DCs inspired by existing therapies.
Collapse
Affiliation(s)
- Patrick Han
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Douglas Hanlon
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Olga Sobolev
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States
| | - Rabib Chaudhury
- Department of Chemical and Environmental Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, United States
| | - Richard L Edelson
- Department of Dermatology, School of Medicine, Yale University, New Haven, CT, United States.
| |
Collapse
|
25
|
Foster JB, Madsen PJ, Hegde M, Ahmed N, Cole KA, Maris JM, Resnick AC, Storm PB, Waanders AJ. Immunotherapy for pediatric brain tumors: past and present. Neuro Oncol 2019; 21:1226-1238. [PMID: 31504801 PMCID: PMC6784275 DOI: 10.1093/neuonc/noz077] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The field of cancer immunotherapy has progressed at an accelerated rate over the past decade. Pediatric brain tumors thus far have presented a formidable challenge for immunotherapy development, given their typically low mutational burden, location behind the blood-brain barrier in a unique tumor microenvironment, and intratumoral heterogeneity. Despite these challenges, recent developments in the field have resulted in exciting preclinical evidence for various immunotherapies and multiple clinical trials. This work reviews the history and advances in active immunotherapy, checkpoint blockade, and adoptive T-cell therapy for pediatric brain tumors, including ongoing clinical trials.
Collapse
Affiliation(s)
- Jessica B Foster
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Peter J Madsen
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Meenakshi Hegde
- Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Nabil Ahmed
- Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Kristina A Cole
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - John M Maris
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Adam C Resnick
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Data Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Phillip B Storm
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Data Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Angela J Waanders
- Division of Oncology, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| |
Collapse
|
26
|
Adhikaree J, Franks HA, Televantos C, Vaghela P, Kaur AP, Walker D, Schmitz M, Jackson AM, Patel PM. Impaired circulating myeloid CD1c+ dendritic cell function in human glioblastoma is restored by p38 inhibition - implications for the next generation of DC vaccines. Oncoimmunology 2019; 8:1593803. [PMID: 31143512 PMCID: PMC6527277 DOI: 10.1080/2162402x.2019.1593803] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/22/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023] Open
Abstract
Current treatments for glioblastoma (GBM) have limited efficacy and significant morbidity and therefore new strategies are urgently needed. Dendritic cells have the power to create anti-tumor immune responses. The greater potency of circulating dendritic cells (DC) over laboratory-generated monocyte-derived DC makes them exciting new immunotherapeutic candidates. To determine the immune status of GBM patients we initially investigated the frequency and function of circulating DC subsets. Furthermore, we tested the therapeutic potential of inhibiting the p38 mitogen-activated protein kinase pathway (p38i) in circulating DC to overcome DC dysfunction. GBM patients (n = 16) had significantly reduced numbers of the major myeloid circulating dendritic cell (cDC2) and plasmacytoid DC vs healthy controls; 1736 vs 4975 (p = 0.028) and 893 vs 2287 cells/mL (P = <0.001) respectively. This inversely correlated with dexamethasone (Dex) dose in a log-linear model, and disease status. Patients' cDC2 were immature with impaired interleukin (IL)-12 secretion, reduced IL-12:IL-10 ratio, and low HLA-DR and CD86 expression. Exposure of healthy donor cDC2 to Dex or GBM cell lysate resulted in a similar low IL-12:IL-10 ratio. Inhibition of p38 restored the IL-12:IL-10 balance in Dex or tumor lysate-conditioned healthy cDC2 and enhanced T-cell proliferation and interferon-gamma (IFNγ) production. Importantly, patient-derived cDC2 showed a similar reversal of DC dysfunction with p38i. This study demonstrates the therapeutic potential of developing the next generation of DC vaccines using enhanced p38i-conditioned cDC2. We will therefore shortly embark on a clinical trial of adoptively transferred, p38 MAPK-inhibited cDC2 in adults with GBM.
Collapse
Affiliation(s)
- Jason Adhikaree
- Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK
| | - Hester Ann Franks
- Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK
| | | | - Poonam Vaghela
- Division of Cancer and Stem Cells, Host-Tumour Interactions Group, UK
| | | | - David Walker
- Children’s Brain Tumour Research Centre, University of Nottingham, Nottingham, UK
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | |
Collapse
|
27
|
McGranahan T, Therkelsen KE, Ahmad S, Nagpal S. Current State of Immunotherapy for Treatment of Glioblastoma. Curr Treat Options Oncol 2019; 20:24. [PMID: 30790064 PMCID: PMC6394457 DOI: 10.1007/s11864-019-0619-4] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OPINION STATEMENT At this time, there are no FDA-approved immune therapies for glioblastoma (GBM) despite many unique therapies currently in clinical trials. GBM is a highly immunosuppressive tumor and there are limitations to a safe immune response in the central nervous system. To date, there have been several failures of phase 3 immune therapy clinical trials in GBM. These trials have targeted single components of an antitumor immune response. Learning from these failures, the future of immunotherapy for GBM appears most hopeful for combination of immune therapies to overcome the profound immunosuppression of this disease. Understanding biomarkers for appropriate patient selection as well as tumor progression are necessary for implementation of immunotherapy for GBM.
Collapse
Affiliation(s)
- Tresa McGranahan
- Department of Neurology, UW Medicine, University of Washington, Seattle, WA USA
| | | | - Sarah Ahmad
- Department of Neurology, Stanford University, Stanford, CA USA
| | - Seema Nagpal
- Department of Neurology, Stanford University, Stanford, CA USA
| |
Collapse
|
28
|
Schiffer D, Annovazzi L, Casalone C, Corona C, Mellai M. Glioblastoma: Microenvironment and Niche Concept. Cancers (Basel) 2018; 11:cancers11010005. [PMID: 30577488 PMCID: PMC6357107 DOI: 10.3390/cancers11010005] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 01/11/2023] Open
Abstract
The niche concept was originally developed to describe the location of normal neural stem cells (NSCs) in the subependymal layer of the sub-ventricular zone. In this paper, its significance has been extended to the location of tumor stem cells in glioblastoma (GB) to discuss the relationship between GB stem cells (GSCs) and endothelial cells (ECs). Their interaction is basically conceived as responsible for tumor growth, invasion and recurrence. Niches are described as the points of utmost expression of the tumor microenvironment (TME), therefore including everything in the tumor except for tumor cells: NSCs, reactive astrocytes, ECs, glioma-associated microglia/macrophages (GAMs), myeloid cells, pericytes, fibroblasts, etc. and all intrinsic and extrinsic signaling pathways. Perivascular (PVNs), perinecrotic (PNNs) and invasive niches were described from the pathological point of view, highlighting the basic significance of the EC/tumor stem cell couple. PNN development was reinterpreted based on the concept that hyperproliferative areas of GB are composed of GSCs/progenitors. TME was depicted in its function as the main regulator of everything that happens in the tumor. A particular emphasis was given to GAMs, pericytes and reactive astrocytes as important elements affecting proliferation, growth, invasion and resistance to therapies of tumor cells.
Collapse
Affiliation(s)
- Davide Schiffer
- Professore Emerito di Neurologia, Università di Torino, Corso Bramante 88/90, 10126 Torino, Italy.
| | - Laura Annovazzi
- Ex Centro Ricerche/Fondazione Policlinico di Monza, Via P. Micca 29, 13100 Vercelli, Italy.
| | - Cristina Casalone
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Via Bologna 148, 10154 Torino, Italy.
| | - Cristiano Corona
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Via Bologna 148, 10154 Torino, Italy.
| | - Marta Mellai
- Dipartimento di Scienze della Salute, Scuola di Medicina, Università del Piemonte Orientale "A. Avogadro", Corso Mazzini 18, 28100 Novara, Italy.
- Fondazione Edo ed Elvo Tempia Valenta-Onlus, Via Malta 3, 13900 Biella, Italy.
| |
Collapse
|
29
|
Nadaradjane A, Briand J, Bougras-Cartron G, Disdero V, Vallette FM, Frenel JS, Cartron PF. miR-370-3p Is a Therapeutic Tool in Anti-glioblastoma Therapy but Is Not an Intratumoral or Cell-free Circulating Biomarker. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 13:642-650. [PMID: 30497054 PMCID: PMC6258828 DOI: 10.1016/j.omtn.2018.09.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022]
Abstract
In the last decade, microRNAs (miRs) have been described as biomarkers and therapeutic agents. Based on this finding, our aim here is to know if (1) miRNA-370-3p can be used as a biomarker associated with a favorable survival and if (2) miRNA-370-3p can be used as a therapeutic tool that increases the efficiency of standard anti-GBM treatment. A first approach using the data available on the “Prognostic miRNA Database” indicated that the expression level of miRNA-370-3p in GBM (T-miR-370-3p) is not associated with a prognosis value for survival. A second approach quantifying the expression level of cell-free circulating miRNA-370-3p (cfc-miR-370-3p) also indicated that cfc-miR-370-3p is not associated with a prognosis value for survival. To investigate whether miR-370-3p can be used in vivo to increase the anti-GBM effect of TMZ, we then used the model of LN18-induced GBMs in mice. Our data indicated that the miRNA-370-3p/TMZ treatment was two times more efficient than the TMZ treatment for decreasing the tumor volume. In addition, our study correlated the decrease of tumor volume induced by the miRNA-370-3p/TMZ treatment with the decrease in FOXM1 and MGMT (i.e., two targets of miR-370-3p). Our data thus support the idea that miR-370-3p could be used as therapeutic tool for anti-glioblastoma therapy, but not as a biomarker.
Collapse
Affiliation(s)
- Arulraj Nadaradjane
- Equipe Apoptose & Progression Tumorale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERM U1232, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain, France; Cancéropole Grand-Ouest, réseau Epigénétique (RepiCGO), Université de Nantes, Nantes, France; EpiSAVMEN Consortium (Région Pays de la Loire), Université de Nantes, Nantes, France
| | - Joséphine Briand
- Equipe Apoptose & Progression Tumorale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERM U1232, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain, France; Cancéropole Grand-Ouest, réseau Epigénétique (RepiCGO), Université de Nantes, Nantes, France; EpiSAVMEN Consortium (Région Pays de la Loire), Université de Nantes, Nantes, France
| | - Gwenola Bougras-Cartron
- Equipe Apoptose & Progression Tumorale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERM U1232, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain, France; Cancéropole Grand-Ouest, réseau Epigénétique (RepiCGO), Université de Nantes, Nantes, France; EpiSAVMEN Consortium (Région Pays de la Loire), Université de Nantes, Nantes, France
| | - Valentine Disdero
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest site René Gauducheau, Saint Herblain, France
| | - François M Vallette
- Equipe Apoptose & Progression Tumorale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERM U1232, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain, France; EpiSAVMEN Consortium (Région Pays de la Loire), Université de Nantes, Nantes, France; LabEX IGO, Université de Nantes, Nantes, France
| | - Jean-Sébastien Frenel
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest site René Gauducheau, Saint Herblain, France
| | - Pierre-François Cartron
- Equipe Apoptose & Progression Tumorale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERM U1232, Nantes, France; Faculté de Médecine, Université de Nantes, Nantes, France; LaBCT, Institut de Cancérologie de l'Ouest, Saint Herblain, France; Cancéropole Grand-Ouest, réseau Epigénétique (RepiCGO), Université de Nantes, Nantes, France; EpiSAVMEN Consortium (Région Pays de la Loire), Université de Nantes, Nantes, France; LabEX IGO, Université de Nantes, Nantes, France.
| |
Collapse
|
30
|
Eagles ME, Nassiri F, Badhiwala JH, Suppiah S, Almenawer SA, Zadeh G, Aldape KD. Dendritic cell vaccines for high-grade gliomas. Ther Clin Risk Manag 2018; 14:1299-1313. [PMID: 30100728 PMCID: PMC6067774 DOI: 10.2147/tcrm.s135865] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common and fatal primary adult brain tumor. To date, various promising chemotherapeutic regimens have been trialed for use in GBM; however, temozolomide (TMZ) therapy remains the only US Food and Drug Administration-approved first-line chemotherapeutic option for newly diagnosed GBM. Despite maximal therapy with surgery and combined concurrent chemoradiation and adjuvant TMZ therapy, the median overall survival remains approximately 14 months. Given the failure of conventional chemotherapeutic strategies in GBM, there has been renewed interest in the role of immunotherapy in GBM. Dendritic cells are immune antigen-presenting cells that play a role in both the innate and adaptive immune system, thereby making them prime vehicles for immunotherapy via dendritic cell vaccinations (DCVs) in various cancers. There is great enthusiasm surrounding the use of DCVs for GBM with multiple ongoing trials. In this review, we comprehensively summarize the safety, efficacy, and quality of life results from 33 trials reporting on DCV for high-grade gliomas.
Collapse
Affiliation(s)
- Matthew E Eagles
- Section of Neurosurgery, Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Farshad Nassiri
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada, .,MacFeeters-Hamilton Neuro-Oncology Program, University Health Network, Toronto, ON, Canada
| | - Jetan H Badhiwala
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada,
| | - Suganth Suppiah
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada,
| | - Saleh A Almenawer
- Division of Neurosurgery, Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Gelareh Zadeh
- MacFeeters-Hamilton Neuro-Oncology Program, University Health Network, Toronto, ON, Canada.,Division of Neurosurgery, University Health Network, Toronto, ON, Canada
| | - Kenneth D Aldape
- MacFeeters-Hamilton Neuro-Oncology Program, University Health Network, Toronto, ON, Canada.,Division of Pathology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
31
|
Healy V, O'Halloran P, O'Brien S, Beausang A, Caird J. CNS metastasis secondary to malignant-mixed Müllerian tumor: case report and review of therapeutics. CNS Oncol 2017; 6:315-323. [PMID: 28990810 DOI: 10.2217/cns-2017-0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This paper reviews CNS involvement secondary to malignant-mixed Müllerian tumor or uterine carcinosarcoma, a rare aggressive biphasic Müllerian tumor. We report a cerebellar metastasis with epithelial and mesenchymal components, demonstrating heterologous rhabdomyogenic and chondroblastic differentiation. The patient had undergone total abdominal hysterectomy and bilateral salpingo-oophorectomy for palliation of symptomatic chemotherapy-resistant node-positive disease. CNS involvement is rare, and prognostically poor, and suggestively poorer in predominantly sarcomatous metastases. Multimodal therapy is indicated; in solitary metastases, surgical resection or stereotactic radiosurgery is included, followed by whole brain radiotherapy. In unresectable brain metastases, stereotactic radiosurgery and whole brain radiotherapy warrant consideration in up to 2-3 metastases. In multiple metastases, palliative steroid therapy or cranial irradiation may be considered. Combination or platinum-based chemotherapy (i.e., ifosfamide-paclitaxel or carboplatin-paclitaxel) is indicated in all stages, with a role in both disease cure and control-directed management. Targeted therapeutics have thus far not demonstrated significant clinical efficacy.
Collapse
Affiliation(s)
- Vincent Healy
- Department of Neurosurgery, National Neurosurgery Centre, Beaumont Hospital, Dublin 11, Ireland
| | - Philip O'Halloran
- Department of Neurosurgery, National Neurosurgery Centre, Beaumont Hospital, Dublin 11, Ireland
| | - Sorca O'Brien
- Department of Gynaecological Oncology, UCD School of Medicine, MMUH, Dublin 7, Ireland
| | - Alan Beausang
- Department of Neuropathology, Beaumont Hospital, Dublin 9, Ireland
| | - John Caird
- Department of Neurosurgery, National Neurosurgery Centre, Beaumont Hospital, Dublin 11, Ireland
| |
Collapse
|
32
|
Advances in Monitoring Cell-Based Therapies with Magnetic Resonance Imaging: Future Perspectives. Int J Mol Sci 2017; 18:ijms18010198. [PMID: 28106829 PMCID: PMC5297829 DOI: 10.3390/ijms18010198] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/05/2017] [Accepted: 01/10/2017] [Indexed: 01/07/2023] Open
Abstract
Cell-based therapies are currently being developed for applications in both regenerative medicine and in oncology. Preclinical, translational, and clinical research on cell-based therapies will benefit tremendously from novel imaging approaches that enable the effective monitoring of the delivery, survival, migration, biodistribution, and integration of transplanted cells. Magnetic resonance imaging (MRI) offers several advantages over other imaging modalities for elucidating the fate of transplanted cells both preclinically and clinically. These advantages include the ability to image transplanted cells longitudinally at high spatial resolution without exposure to ionizing radiation, and the possibility to co-register anatomical structures with molecular processes and functional changes. However, since cellular MRI is still in its infancy, it currently faces a number of challenges, which provide avenues for future research and development. In this review, we describe the basic principle of cell-tracking with MRI; explain the different approaches currently used to monitor cell-based therapies; describe currently available MRI contrast generation mechanisms and strategies for monitoring transplanted cells; discuss some of the challenges in tracking transplanted cells; and suggest future research directions.
Collapse
|