1
|
Gaoual Y, Mahyaoui A, Yachi L, Bouatia M, Aliat Z, Rahali Y. Advancements and challenges in CAR T cell therapy for pediatric brain tumors: A review. J Oncol Pharm Pract 2025:10781552251331609. [PMID: 40156311 DOI: 10.1177/10781552251331609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy represents a groundbreaking advancement in immunotherapy, initially gaining FDA approval for treating hematological malignancies. This therapy has shown promising results in solid tumors, particularly in pediatric brain tumors, which are the leading cause of cancer-related death in children. CAR T cells are engineered to target specific antigens on tumor cells, thereby reducing off-target effects and increasing the cytotoxic impact on cancer cells. Over the years, CAR T cell technology has evolved through five generations, each enhancing the structure, functionality, and safety of these cells. Despite these advancements, the application of CAR T cells in solid tumors, especially within the central nervous system (CNS), faces significant challenges. These include the physical barrier posed by the blood-brain barrier (BBB), the immunosuppressive tumor microenvironment (TME), and the heterogeneity of tumor antigens. The review discusses several promising antigenic targets for CAR T cells in pediatric brain tumors, such as HER2, EphA2, IL-13Rα2, and Survivin, which have been explored in recent clinical trials. These trials have shown early promise in improving patient outcomes, though the risks of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) remain concerns. The future of CAR T cell therapy lies in overcoming these barriers through innovative approaches like "Armored CARs" or TRUCKs, designed to modulate the TME and improve CAR T cell efficacy in solid tumors. Additionally, combination therapies and safety switches in next-generation CAR T cells are being explored to enhance therapeutic potential while minimizing adverse effects.
Collapse
Affiliation(s)
- Yasmina Gaoual
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
| | - Adam Mahyaoui
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
| | - Lamyae Yachi
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Children's hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of analytical chemistry and bromatology, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| | - Mustapha Bouatia
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Children's hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of analytical chemistry and bromatology, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
- Ibn Sina University Hospital Center, 10 170 Rabat, Morocco
| | - Zineb Aliat
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| | - Younes Rahali
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10 170 Rabat, Morocco
- Specialties Hospital of Rabat, Ibn Sina University Hospital, 10 170 Rabat, Morocco
- Ibn Sina University Hospital Center, 10 170 Rabat, Morocco
- Team of Formulation and Quality Control of Health Products, Faculty of Medicine and Pharmacy, Mohammed V University- Rabat, 10 170 Rabat, Morocco
| |
Collapse
|
2
|
Giordano G, Tucciarello C, Merlini A, Cutrupi S, Pignochino Y. Targeting the EphA2 pathway: could it be the way for bone sarcomas? Cell Commun Signal 2024; 22:433. [PMID: 39252029 PMCID: PMC11382444 DOI: 10.1186/s12964-024-01811-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Bone sarcomas are malignant tumors of mesenchymal origin. Complete surgical resection is the cornerstone of multidisciplinary treatment. However, advanced, unresectable forms remain incurable. A crucial step towards addressing this challenge involves comprehending the molecular mechanisms underpinning tumor progression and metastasis, laying the groundwork for innovative precision medicine-based interventions. We previously showed that tyrosine kinase receptor Ephrin Type-A Receptor 2 (EphA2) is overexpressed in bone sarcomas. EphA2 is a key oncofetal protein implicated in metastasis, self-renewal, and chemoresistance. Molecular, genetic, biochemical, and pharmacological approaches have been developed to target EphA2 and its signaling pathway aiming to interfere with its tumor-promoting effects or as a carrier for drug delivery. This review synthesizes the main functions of EphA2 and their relevance in bone sarcomas, providing strategies devised to leverage this receptor for diagnostic and therapeutic purposes, with a focus on its applicability in the three most common bone sarcoma histotypes: osteosarcoma, chondrosarcoma, and Ewing sarcoma.
Collapse
Affiliation(s)
- Giorgia Giordano
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Cristina Tucciarello
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Ymera Pignochino
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy.
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy.
| |
Collapse
|
3
|
Bright RK. Preclinical support for tumor protein D52 as a cancer vaccine antigen. Hum Vaccin Immunother 2023; 19:2273699. [PMID: 37904517 PMCID: PMC10760363 DOI: 10.1080/21645515.2023.2273699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/18/2023] [Indexed: 11/01/2023] Open
Abstract
Overexpressed tumor-associated antigens (TAAs) are a large group that includes proteins found at increased levels in tumors compared to healthy cells. Universal tumor expression can be defined as overexpression in all cancers examined as has been shown for Tumor Protein D52. TPD52 is an over expressed TAA actively involved in transformation, leading to increased proliferation and metastasis. TPD52 overexpression has been demonstrated in many human adult and pediatric malignancies. The murine orthologue of TPD52 (mD52) parallels normal tissue expression patterns and known functions of human TPD52 (hD52). Here in we present our preclinical studies over the past 15 years which have demonstrated that vaccine induced immunity against mD52 is effective against multiple cancers in murine models, without inducing autoimmunity against healthy tissues and cells.
Collapse
Affiliation(s)
- Robert K. Bright
- Department of Immunology and Molecular Microbiology, School of Medicine and Cancer Center, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
4
|
Guzman G, Pellot K, Reed MR, Rodriguez A. CAR T-cells to treat brain tumors. Brain Res Bull 2023; 196:76-98. [PMID: 36841424 DOI: 10.1016/j.brainresbull.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/18/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
Tremendous success using CAR T therapy in hematological malignancies has garnered significant interest in developing such treatments for solid tumors, including brain tumors. This success, however, has yet to be mirrored in solid organ neoplasms. CAR T function has shown limited efficacy against brain tumors due to several factors including the immunosuppressive tumor microenvironment, blood-brain barrier, and tumor-antigen heterogeneity. Despite these considerations, CAR T-cell therapy has the potential to be implemented as a treatment modality for brain tumors. Here, we review adult and pediatric brain tumors, including glioblastoma, diffuse midline gliomas, and medulloblastomas that continue to portend a grim prognosis. We describe insights gained from different preclinical models using CAR T therapy against various brain tumors and results gathered from ongoing clinical trials. Furthermore, we outline the challenges limiting CAR T therapy success against brain tumors and summarize advancements made to overcome these obstacles.
Collapse
Affiliation(s)
- Grace Guzman
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Megan R Reed
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
5
|
Zhao S, Li J, Zhang H, Qi L, Du Y, Kogiso M, Braun FK, Xiao S, Huang Y, Li J, Teo WY, Lindsay H, Baxter P, Su JMF, Adesina A, Laczik M, Genevini P, Veillard AC, Schvartzman S, Berguet G, Ding SR, Du L, Stephan C, Yang J, Davies PJA, Lu X, Chintagumpala M, Parsons DW, Perlaky L, Xia YF, Man TK, Huang Y, Sun D, Li XN. Epigenetic Alterations of Repeated Relapses in Patient-matched Childhood Ependymomas. Nat Commun 2022; 13:6689. [PMID: 36335125 PMCID: PMC9637194 DOI: 10.1038/s41467-022-34514-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2022] Open
Abstract
Recurrence is frequent in pediatric ependymoma (EPN). Our longitudinal integrated analysis of 30 patient-matched repeated relapses (3.67 ± 1.76 times) over 13 years (5.8 ± 3.8) reveals stable molecular subtypes (RELA and PFA) and convergent DNA methylation reprogramming during serial relapses accompanied by increased orthotopic patient derived xenograft (PDX) (13/27) formation in the late recurrences. A set of differentially methylated CpGs (DMCs) and DNA methylation regions (DMRs) are found to persist in primary and relapse tumors (potential driver DMCs) and are acquired exclusively in the relapses (potential booster DMCs). Integrating with RNAseq reveals differentially expressed genes regulated by potential driver DMRs (CACNA1H, SLC12A7, RARA in RELA and HSPB8, GMPR, ITGB4 in PFA) and potential booster DMRs (PLEKHG1 in RELA and NOTCH, EPHA2, SUFU, FOXJ1 in PFA tumors). DMCs predicators of relapse are also identified in the primary tumors. This study provides a high-resolution epigenetic roadmap of serial EPN relapses and 13 orthotopic PDX models to facilitate biological and preclinical studies.
Collapse
Affiliation(s)
- Sibo Zhao
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.413584.f0000 0004 0383 5679Jane and John Justin Neurosciences Center, Cook Children’s Medical Center, Fort Worth, TX 76104 USA ,grid.413584.f0000 0004 0383 5679Hematology and Oncology Center, Cook Children’s Medical Center, Fort Worth, TX 76104 USA
| | - Jia Li
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA ,grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA ,grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University; and Guangzhou Laboratory, Bioland, 510120 Guangzhou, Guangdong P. R. China
| | - Huiyuan Zhang
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Lin Qi
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Yuchen Du
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Mari Kogiso
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Frank K. Braun
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Sophie Xiao
- grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Yulun Huang
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.263761.70000 0001 0198 0694Department of Neurosurgery and Brain and Nerve Research Laboratory, the First Affiliated Hospital, and Department of Neurosurgery, Dushu Lake Hospital, Suzhou Medical College, Soochow University, 215007 Suzhou, P. R. China
| | - Jianfang Li
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Wan-Yee Teo
- grid.410724.40000 0004 0620 9745Humphrey Oei Institute of Cancer Research, National Cancer Center Singapore, Singapore, 169610 Singapore ,grid.428397.30000 0004 0385 0924Cancer and Stem Cell Biology Program, Duke-NUS Medical School Singapore, Singapore, Singapore ,grid.414963.d0000 0000 8958 3388KK Women’s & Children’s Hospital Singapore, Singapore, Singapore ,grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Holly Lindsay
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Patricia Baxter
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Jack M. F. Su
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Adekunle Adesina
- grid.39382.330000 0001 2160 926XDepartment of Pathology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Miklós Laczik
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Paola Genevini
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | | | - Sol Schvartzman
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Geoffrey Berguet
- grid.424287.f0000 0004 0555 845XEpigenetic Services, Diagenode, Liège Belgium
| | - Shi-Rong Ding
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Department of Radiation, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong P. R. China
| | - Liping Du
- grid.16753.360000 0001 2299 3507Clinical Cytogenetic Laboratory, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Clifford Stephan
- grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA
| | - Jianhua Yang
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Peter J. A. Davies
- grid.264756.40000 0004 4687 2082Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030 USA
| | - Xinyan Lu
- grid.16753.360000 0001 2299 3507Clinical Cytogenetic Laboratory, Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Murali Chintagumpala
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Donald William Parsons
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Laszlo Perlaky
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yun-Fei Xia
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Department of Radiation, Sun Yat-sen University Cancer Center, 510060 Guangzhou, Guangdong P. R. China
| | - Tsz-Kwong Man
- grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA
| | - Yun Huang
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Deqiang Sun
- grid.264756.40000 0004 4687 2082Center for Epigenetics & Disease Prevention, Texas A&M University, Houston, TX 77030 USA
| | - Xiao-Nan Li
- grid.39382.330000 0001 2160 926XPre-clinical Neuro-oncology Research Program, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XTexas Children’s Cancer Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030 USA ,grid.16753.360000 0001 2299 3507Program of Precision Medicine PDOX Modeling of Pediatric Tumors, Division of Hematology-Oncology, Neuro-Oncology & Stem Cell transplantation, Ann & Robert H. Lurie Children’s Hospital of Chicago; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
6
|
Day CA, Hinchcliffe EH, Robinson JP. H3K27me3 in Diffuse Midline Glioma and Epithelial Ovarian Cancer: Opposing Epigenetic Changes Leading to the Same Poor Outcomes. Cells 2022; 11:cells11213376. [PMID: 36359771 PMCID: PMC9655269 DOI: 10.3390/cells11213376] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/29/2022] Open
Abstract
Histone post-translational modifications modulate gene expression through epigenetic gene regulation. The core histone H3 family members, H3.1, H3.2, and H3.3, play a central role in epigenetics. H3 histones can acquire many post-translational modifications, including the trimethylation of H3K27 (H3K27me3), which represses transcription. Triple methylation of H3K27 is performed by the histone methyltransferase Enhancer of Zeste Homologue 2 (EZH2), a component of the Polycomb Repressive Complex 2. Both global increases and decreases in H3K27me3 have been implicated in a wide range of cancer types. Here, we explore how opposing changes in H3K27me3 contribute to cancer by highlighting its role in two vastly different cancer types; (1) a form of glioma known as diffuse midline glioma H3K27-altered and (2) epithelial ovarian cancer. These two cancers vary widely in the age of onset, sex, associated mutations, and cell and organ type. However, both diffuse midline glioma and ovarian cancer have dysregulation of H3K27 methylation, triggering changes to the cancer cell transcriptome. In diffuse midline glioma, the loss of H3K27 methylation is a primary driving factor in tumorigenesis that promotes glial cell stemness and silences tumor suppressor genes. Conversely, hypermethylation of H3K27 occurs in late-stage epithelial ovarian cancer, which promotes tumor vascularization and tumor cell migration. By using each cancer type as a case study, this review emphasizes the importance of H3K27me3 in cancer while demonstrating that the mechanisms of histone H3 modification and subsequent gene expression changes are not a one-size-fits-all across cancer types.
Collapse
Affiliation(s)
- Charles A. Day
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Mayo Clinic, Rochester, MN 55902, USA
- Correspondence:
| | - Edward H. Hinchcliffe
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - James P. Robinson
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
7
|
Lin YJ, Mashouf LA, Lim M. CAR T Cell Therapy in Primary Brain Tumors: Current Investigations and the Future. Front Immunol 2022; 13:817296. [PMID: 35265074 PMCID: PMC8899093 DOI: 10.3389/fimmu.2022.817296] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/20/2022] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor T cells (CAR T cells) are engineered cells expressing a chimeric antigen receptor (CAR) against a specific tumor antigen (TA) that allows for the identification and elimination of cancer cells. The remarkable clinical effect seen with CAR T cell therapies against hematological malignancies have attracted interest in developing such therapies for solid tumors, including brain tumors. Glioblastoma (GBM) is the most common primary brain tumor in adults and is associated with poor prognosis due to its highly aggressive nature. Pediatric brain cancers are similarly aggressive and thus are a major cause of pediatric cancer-related death. CAR T cell therapy represents a promising avenue for therapy against these malignancies. Several specific TAs, such as EGFR/EGFRvIII, IL13Rα2, B7-H3, and HER2, have been targeted in preclinical studies and clinical trials. Unfortunately, CAR T cells against brain tumors have showed limited efficacy due to TA heterogeneity, difficulty trafficking from blood to tumor sites, and the immunosuppressive tumor microenvironment. Here, we review current CAR T cell approaches in treating cancers, with particular focus on brain cancers. We also describe a novel technique of focused ultrasound controlling the activation of engineered CAR T cells to achieve the safer cell therapies. Finally, we summarize the development of combinational strategies to improve the efficacy and overcome historical limitations of CAR T cell therapy.
Collapse
Affiliation(s)
- Ya-Jui Lin
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, United States.,Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Leila A Mashouf
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, United States.,Harvard Medical School, Boston, MA, United States
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, United States
| |
Collapse
|
8
|
Pediatric glioblastoma: mechanisms of immune evasion and potential therapeutic opportunities. Cancer Immunol Immunother 2022; 71:1813-1822. [PMID: 35020009 DOI: 10.1007/s00262-021-03131-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
Pediatric glioblastoma is relatively rare compared with its adult counterpart but is associated with a similarly grim prognosis. Available data indicate that pediatric glioblastomas are molecularly distinct from adult tumors, and relatively little is known about the pediatric glioblastoma tumor microenvironment (TME). Cancer immunotherapy has emerged as a new pillar of cancer treatment and is revolutionizing the care of patients with many advanced solid tumors, including melanoma, non-small cell lung cancer, head and neck cancer, and renal cell carcinoma. Unfortunately, attempts to treat adult glioblastoma with current immunotherapies have had limited success to date. Nevertheless, the immune milieu in pediatric glioblastoma is distinct from that found in adult tumors, and evidence suggests that pediatric tumors are less immunosuppressive. As a result, immunotherapies should be specifically evaluated in the pediatric context. The purpose of this review is to explore known and emerging mechanisms of immune evasion in pediatric glioblastoma and highlight potential opportunities for implementing immunotherapy in the treatment of these devastating pediatric brain tumors.
Collapse
|
9
|
D'Orazi G, Cordani M, Cirone M. Oncogenic pathways activated by pro-inflammatory cytokines promote mutant p53 stability: clue for novel anticancer therapies. Cell Mol Life Sci 2021; 78:1853-1860. [PMID: 33070220 PMCID: PMC11072129 DOI: 10.1007/s00018-020-03677-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/03/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022]
Abstract
Inflammation and cancerogenesis are strongly interconnected processes, not only because inflammation promotes DNA instability, but also because both processes are driven by pathways such as NF-kB, STAT3, mTOR and MAPKs. Interestingly, these pathways regulate the release of pro-inflammatory cytokines such as IL-6, TNF-α and IL-1β that in turn control their activation and play a crucial role in shaping immune response. The transcription factor p53 is the major tumor suppressor that is often mutated in cancer, contributing to tumor progression. In this overview, we highlight how the interplay between pro-inflammatory cytokines and pro-inflammatory/pro-oncogenic pathways, regulating and being regulated by UPR signaling and autophagy, affects the stability of mutp53 that in turn is able to control autophagy, UPR signaling, cytokine release and the activation of the same oncogenic pathways to preserve its own stability and promote tumorigenesis. Interrupting these positive feedback loops may represent a promising strategy in anticancer therapy, particularly against cancers carrying mutp53.
Collapse
Affiliation(s)
- Gabriella D'Orazi
- Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Cordani
- IMDEA Nanociencia, C/Faraday 9, Ciudad Universitaria de Cantoblanco, Madrid, Spain
| | - Mara Cirone
- Department of Experimental Medicine, Laboratory Affiliated to Pasteur Institute Italy Foundation Cenci Bolognetti, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
10
|
Wu WT, Lin WY, Chen YW, Lin CF, Wang HH, Wu SH, Lee YY. New Era of Immunotherapy in Pediatric Brain Tumors: Chimeric Antigen Receptor T-Cell Therapy. Int J Mol Sci 2021; 22:2404. [PMID: 33673696 PMCID: PMC7957810 DOI: 10.3390/ijms22052404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/16/2021] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy, including chimeric antigen receptor (CAR) T-cell therapy, immune checkpoint inhibitors, cancer vaccines, and dendritic cell therapy, has been incorporated as a fifth modality of modern cancer care, along with surgery, radiation, chemotherapy, and target therapy. Among them, CAR T-cell therapy emerges as one of the most promising treatments. In 2017, the first two CAR T-cell drugs, tisagenlecleucel and axicabtagene ciloleucel for B-cell acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL), respectively, were approved by the Food and Drug Administration (FDA). In addition to the successful applications to hematological malignancies, CAR T-cell therapy has been investigated to potentially treat solid tumors, including pediatric brain tumor, which serves as the leading cause of cancer-associated death for children and adolescents. However, the employment of CAR T-cell therapy in pediatric brain tumors still faces multiple challenges, such as CAR T-cell transportation and expansion through the blood-brain barrier, and identification of the specific target antigen on the tumor surface and immunosuppressive tumor microenvironment. Nevertheless, encouraging outcomes in both clinical and preclinical trials are coming to light. In this article, we outline the current propitious progress and discuss the obstacles needed to be overcome in order to unveil a new era of treatment in pediatric brain tumors.
Collapse
Affiliation(s)
- Wan-Tai Wu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (W.-T.W.); (Y.-W.C.); (C.-F.L.)
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei 112201, Taiwan
| | - Wen-Ying Lin
- Department of Internal Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
| | - Yi-Wei Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (W.-T.W.); (Y.-W.C.); (C.-F.L.)
- Division of Radiation Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Chun-Fu Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (W.-T.W.); (Y.-W.C.); (C.-F.L.)
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Hsin-Hui Wang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Department of Pediatrics, Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Szu-Hsien Wu
- Department of Plastic and Reconstructive Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan;
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Yen Lee
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (W.-T.W.); (Y.-W.C.); (C.-F.L.)
- Division of Pediatric Neurosurgery, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 201, Section 2, Shih-Pai Road, Taipei 112201, Taiwan
- Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| |
Collapse
|
11
|
Patterson JD, Henson JC, Breese RO, Bielamowicz KJ, Rodriguez A. CAR T Cell Therapy for Pediatric Brain Tumors. Front Oncol 2020; 10:1582. [PMID: 32903405 PMCID: PMC7435009 DOI: 10.3389/fonc.2020.01582] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/22/2020] [Indexed: 12/31/2022] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy has recently begun to be used for solid tumors such as glioblastoma multiforme. Many children with pediatric malignant brain tumors develop extensive long-term morbidity of intensive multimodal curative treatment. Others with certain diagnoses and relapsed disease continue to have limited therapies and a dismal prognosis. Novel treatments such as CAR T cells could potentially improve outcomes and ameliorate the toxicity of current treatment. In this review, we discuss the potential of using CAR therapy for pediatric brain tumors. The emerging insights on the molecular subtypes and tumor microenvironment of these tumors provide avenues to devise strategies for CAR T cell therapy. Unique characteristics of these brain tumors, such as location and associated morbid treatment induced neuro-inflammation, are novel challenges not commonly encountered in adult brain tumors. Despite these considerations, CAR T cell therapy has the potential to be integrated into treatment schema for aggressive pediatric malignant brain tumors in the future.
Collapse
Affiliation(s)
- John D Patterson
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jeffrey C Henson
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Rebecca O Breese
- Department of General Surgery, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States
| | - Kevin J Bielamowicz
- Division of Hematology/Oncology, Department of Pediatrics, Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
12
|
Fang D, Gan H, Cheng L, Lee JH, Zhou H, Sarkaria JN, Daniels DJ, Zhang Z. H3.3K27M mutant proteins reprogram epigenome by sequestering the PRC2 complex to poised enhancers. eLife 2018; 7:36696. [PMID: 29932419 PMCID: PMC6033537 DOI: 10.7554/elife.36696] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/21/2018] [Indexed: 11/21/2022] Open
Abstract
Expression of histone H3.3K27M mutant proteins in human diffuse intrinsic pontine glioma (DIPG) results in a global reduction of tri-methylation of H3K27 (H3K27me3), and paradoxically, H3K27me3 peaks remain at hundreds of genomic loci, a dichotomous change that lacks mechanistic insights. Here, we show that the PRC2 complex is sequestered at poised enhancers, but not at active promoters with high levels of H3.3K27M proteins, thereby contributing to the global reduction of H3K27me3. Moreover, the levels of H3.3K27M proteins are low at the retained H3K27me3 peaks and consequently having minimal effects on the PRC2 activity at these loci. H3K27me3-mediated silencing at specific tumor suppressor genes, including Wilms Tumor 1, promotes proliferation of DIPG cells. These results support a model in which the PRC2 complex is redistributed to poised enhancers in H3.3K27M mutant cells and contributes to tumorigenesis in part by locally enhancing H3K27me3, and hence silencing of tumor suppressor genes.
Collapse
Affiliation(s)
- Dong Fang
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, United States
| | - Haiyun Gan
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, United States
| | - Liang Cheng
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Jeong-Heon Lee
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, United States
| | - Hui Zhou
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, United States
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, United States
| | - David J Daniels
- Department of Neurosurgery, Mayo Clinic, Rochester, United States
| | - Zhiguo Zhang
- Department of Pediatrics and Department of Genetics and Development, Institute for Cancer Genetics, Irving Cancer Research Center, Columbia University, New York, United States
| |
Collapse
|
13
|
Ramsawhook A, Ruzov A, Coyle B. Wilms' Tumor Protein 1 and Enzymatic Oxidation of 5-Methylcytosine in Brain Tumors: Potential Perspectives. Front Cell Dev Biol 2018; 6:26. [PMID: 29623275 PMCID: PMC5874295 DOI: 10.3389/fcell.2018.00026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/05/2018] [Indexed: 12/24/2022] Open
Abstract
The patterns of 5-methylcytosine (5mC) and its oxidized derivatives, 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine (5caC) are reportedly altered in a range of cancers. Likewise, Wilms' Tumor protein 1 (WT1), a transcription factor essential for urogenital, epicardium, and kidney development exhibits aberrant expression in multiple tumors. Interestingly, WT1 directly interacts with TET proteins that catalyze the enzymatic oxidation of 5mC and exhibits high affinity for 5caC-containing DNA substrates in vitro. Here we review recent developments in the fields of Tet-dependent 5mC oxidation and WT1 biology and explore potential perspectives for studying the interplay between TETs and WT1 in brain tumors.
Collapse
Affiliation(s)
- Ashley Ramsawhook
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Alexey Ruzov
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Beth Coyle
- Children's Brain Tumour Research Centre, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
14
|
Madhankumar AB, Mrowczynski OD, Slagle-Webb B, Ravi V, Bourcier AJ, Payne R, Harbaugh KS, Rizk E, Connor JR. Tumor targeted delivery of doxorubicin in malignant peripheral nerve sheath tumors. PLoS One 2018; 13:e0181529. [PMID: 29304038 PMCID: PMC5755733 DOI: 10.1371/journal.pone.0181529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 07/03/2017] [Indexed: 01/22/2023] Open
Abstract
Peripheral nerve sheath tumors are benign tumors that have the potential to transform into malignant peripheral nerve sheath tumors (MPNSTs). Interleukin-13 receptor alpha 2 (IL13Rα2) is a cancer associated receptor expressed in glioblastoma and other invasive cancers. We analyzed IL13Rα2 expression in several MPNST cell lines including the STS26T cell line, as well as in several peripheral nerve sheath tumors to utilize the IL13Rα2 receptor as a target for therapy. In our studies, we demonstrated the selective expression of IL13Rα2 in several peripheral nerve sheath tumors by immunohistochemistry (IHC) and immunoblots. We established a sciatic nerve MPNST mouse model in NIH III nude mice using a luciferase transfected STS26T MPNST cell line. Similarly, analysis of the mouse sciatic nerves after tumor induction revealed significant expression of IL13Rα2 by IHC when compared to a normal sciatic nerve. IL13 conjugated liposomal doxorubicin was formulated and shown to bind and internalized in the MPNST cell culture model demonstrating cytotoxic effect. Our subsequent in vivo investigation in the STS26T MPNST sciatic nerve tumor model indicated that IL13 conjugated liposomal doxorubicin (IL13LIPDXR) was more effective in inhibiting tumor progression compared to unconjugated liposomal doxorubicin (LIPDXR). This further supports that IL13 receptor targeted nanoliposomes is a potential approach for treating MPNSTs.
Collapse
Affiliation(s)
- A. B. Madhankumar
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
- * E-mail:
| | - Oliver D. Mrowczynski
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Becky Slagle-Webb
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Vagisha Ravi
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Alexandre J. Bourcier
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Russell Payne
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Kimberly S. Harbaugh
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - Elias Rizk
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| | - James R. Connor
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA, United States of America
| |
Collapse
|
15
|
Abstract
Ependymoma is a locally aggressive tumor with metastatic potential that arises in diverse locations throughout the brain and spine in children. Tumor and treatment may result in significant morbidity. Cure remains elusive for many patients owing to diverse biology and resistance to conventional therapy. The implementation of systematic postoperative irradiation in clinical trials during the past 20 years has increased the proportion of patients achieving durable disease control with excellent results, as measured by objective functional outcome measures. Clinical, pathologic, and molecular risk stratification should be used to refine treatment regimens for children with ependymoma to reduce the risk of complications associated with therapy and increase the rate of disease control in the setting of combined modality or more intensive therapy. This review covers standards of care and current clinical trials for children with ependymoma, emphasizing the history and evolution of treatment regimens during the past 20 years and the clinical questions they hoped to address.
Collapse
|
16
|
Furtado AD, Ceschin R, Blüml S, Mason G, Jakacki RI, Okada H, Pollack IF, Panigrahy A. Neuroimaging of Peptide-based Vaccine Therapy in Pediatric Brain Tumors: Initial Experience. Neuroimaging Clin N Am 2017; 27:155-166. [PMID: 27889021 DOI: 10.1016/j.nic.2016.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The potential benefits of peptide-based immunotherapy for pediatric brain tumors are under investigation. Treatment-related heterogeneity has resulted in radiographic challenges, including pseudoprogression. Conventional MR imaging has limitations in assessment of different forms of treatment-related heterogeneity, particularly regarding distinguishing true tumor progression from efficacious treatment responses. Advanced neuroimaging techniques, including diffusion magnetic resonance (MR), perfusion MR, and MR spectroscopy, may add value in the assessment of treatment-related heterogeneity. Observations suggest that recent delineation of specific response criteria for immunotherapy of adult brain tumors is likely relevant to the pediatric population and further validation in multicenter pediatric brain tumor peptide-based vaccine studies is warranted.
Collapse
Affiliation(s)
- Andre D Furtado
- Department of Radiology, University of Pittsburgh, 3600 Forbes Avenue, Pittsburgh, PA 15213, USA
| | - Rafael Ceschin
- Department of Radiology, University of Pittsburgh, 3600 Forbes Avenue, Pittsburgh, PA 15213, USA; Department of Bioinformatics, University of Pittsburgh, 5607 Baum Boulevard, Suite 500, Pittsburgh, PA 15206, USA
| | - Stefan Blüml
- Department of Radiology, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, USA
| | - Gary Mason
- Department of Pediatrics, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Regina I Jakacki
- Department of Pediatrics, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco, 505 Parnassus Avenue, M-779, San Francisco, CA 94143, USA
| | - Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Children's Hospital of Pittsburgh, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Ashok Panigrahy
- Department of Radiology, University of Pittsburgh, 3600 Forbes Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
17
|
Varughese RK, Torp SH. Survivin and gliomas: A literature review. Oncol Lett 2016; 12:1679-1686. [PMID: 27588117 PMCID: PMC4998142 DOI: 10.3892/ol.2016.4867] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 05/10/2016] [Indexed: 01/21/2023] Open
Abstract
Gliomas are the most common primary brain tumor, the diagnosis of which is challenging. In this respect, the use of immunohistochemical proliferation markers may aid diagnosis; survivin, also known as Baculoviral IAP Repeat Containing 5, is one such marker. Survivin is a unique member of the inhibitors of apoptosis protein gene family, and is known for its dual function as an apoptosis inhibitor and mitosis regulator. Furthermore, survivin has been demonstrated to be overexpressed in a number of malignancies. The purpose of the present literature review was to gain an overview of studies published on the diagnostic and/or prognostic use of survivin in gliomas. Using PubMed, 19 studies matching the inclusion criteria were ultimately included in the present review. The majority of the studies identified revealed that survivin was significantly associated with other proliferation markers, histological malignancy grade, and inversely associated with prognosis. However, there were a number of inconsistencies between studies, which suggests a requirement for standardization of immunohistochemical procedures.
Collapse
Affiliation(s)
- Rosilin Kotakkathu Varughese
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), 7030 Trondheim, Norway
| | - Sverre Helge Torp
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), 7030 Trondheim, Norway
- Department of Pathology and Medical Genetics, St. Olavs Hospital, 7006 Trondheim, Norway
| |
Collapse
|
18
|
Abstract
Vaccination against cancer-associated antigens has long held the promise of inducting potent antitumor immunity, targeted cytotoxicity while sparing normal tissues, and long-lasting immunologic memory that can provide surveillance against tumor recurrence. Evaluation of vaccination strategies in preclinical brain tumor models has borne out the capacity for the immune system to effectively and safely eradicate established tumors within the central nervous system. Early phase clinical trials have established the feasibility, safety, and immunogenicity of several vaccine platforms, predominantly in patients with glioblastoma. Definitive demonstration of clinical benefit awaits further study, but initial results have been encouraging. With increased understanding of the stimulatory and regulatory pathways that govern immunologic responses and the enhanced capacity to identify novel antigenic targets using genomic interrogation of tumor cells, vaccination platforms for patients with malignant brain tumors are advancing with increasing personalized complexity and integration into combinatorial treatment paradigms.
Collapse
Affiliation(s)
- John H Sampson
- Preston Robert Tisch Brain Tumor Center at Duke, Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida (D.A.M.)
| | - Duane A Mitchell
- Preston Robert Tisch Brain Tumor Center at Duke, Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Department of Neurosurgery, McKnight Brain Institute, University of Florida, Gainesville, Florida (D.A.M.)
| |
Collapse
|
19
|
Abstract
Overexpressed tumor-self antigens represent the largest group of candidate vaccine targets. Those exhibiting a role in oncogenesis may be some of the least studied but perhaps most promising. This review considers this subset of self antigens by highlighting vaccine efforts for some of the better known members and focusing on TPD52, a new promising vaccine target. We shed light on the importance of both preclinical and clinical vaccine studies demonstrating that tolerance and autoimmunity (presumed to preclude this class of antigens from vaccine development) can be overcome and do not present the obstacle that might have been expected. The potential of this class of antigens for broad application is considered, possibly in the context of low tumor burden or adjuvant therapy, as is the need to understand mechanisms of tolerance that are relatively understudied.
Collapse
Key Words
- ALK, Anaplastic lymphoma kinase
- AR, androgen receptor
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte-associated antigen 4
- HLA, human leukocyte antigen
- Her-2/neu, human epithelial growth factor receptor 2
- ODN, oligodeoxynucleotide
- Overexpressed tumor-self antigen
- TAA, tumor associated antigen
- TPD52
- TRAMP, Transgenic adenocarcinoma of the mouse prostate
- Treg, T regulatory cell
- VEGFR2, vascular endothelial growth factor receptor 2
- WT-1, Wilms tumor-1
- hD52
- hD52, human TPD52
- mD52
- mD52, murine TPD52
- oncogenic
- shared
- tumor protein D52
- universal
- vaccine
Collapse
Affiliation(s)
- Robert K Bright
- a Department of Immunology and Molecular Microbiology and the TTUHSC Cancer Center ; Texas Tech University Health Sciences Center ; Lubbock , TX USA
| | | | | |
Collapse
|
20
|
Karsy M, Guan J, Sivakumar W, Neil JA, Schmidt MH, Mahan MA. The genetic basis of intradural spinal tumors and its impact on clinical treatment. Neurosurg Focus 2015; 39:E3. [DOI: 10.3171/2015.5.focus15143] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetic alterations in the cells of intradural spinal tumors can have a significant impact on the treatment options, counseling, and prognosis for patients. Although surgery is the primary therapy for most intradural tumors, radiochemothera-peutic modalities and targeted interventions play an ever-evolving role in treating aggressive cancers and in addressing cancer recurrence in long-term survivors. Recent studies have helped delineate specific genetic and molecular differences between intradural spinal tumors and their intracranial counterparts and have also identified significant variation in therapeutic effects on these tumors. This review discusses the genetic and molecular alterations in the most common intradural spinal tumors in both adult and pediatrie patients, including nerve sheath tumors (that is, neurofibroma and schwannoma), meningioma, ependymoma, astrocytoma (that is, low-grade glioma, anaplastic astrocytoma, and glioblastoma), hemangioblastoma, and medulloblastoma. It also examines the genetics of metastatic tumors to the spinal cord, arising either from the CNS or from systemic sources. Importantly, the impact of this knowledge on therapeutic options and its application to clinical practice are discussed.
Collapse
|
21
|
WT1 Expression in Circulating RNA as a Minimal Residual Disease Marker for AML Patients After Stem-Cell Transplantation. Mol Diagn Ther 2015; 19:205-12. [DOI: 10.1007/s40291-015-0147-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
22
|
Abstract
Background The majority of genetic biomarkers for human cancers are defined by statistical screening of high-throughput genomics data. While a large number of genetic biomarkers have been proposed for diagnostic and prognostic applications, only a small number have been applied in the clinic. Similarly, the use of proteomics methods for the discovery of cancer biomarkers is increasing. The emerging field of proteogenomics seeks to enrich the value of genomics and proteomics approaches by studying the intersection of genomics and proteomics data. This task is challenging due to the complex nature of transcriptional and translation regulatory mechanisms and the disparities between genomic and proteomic data from the same samples. In this study, we have examined tumor antigens as potential biomarkers for breast cancer using genomics and proteomics data from previously reported laser capture microdissected ER+ tumor samples. Results We applied proteogenomic analyses to study the genetic aberrations of 32 tumor antigens determined in the proteomic data. We found that tumor antigens that are aberrantly expressed at the genetic level and expressed at the protein level, are likely involved in perturbing pathways directly linked to the hallmarks of cancer. The results found by proteogenomic analysis of the 32 tumor antigens studied here, capture largely the same pathway irregularities as those elucidated from large-scale screening of genomics analyses, where several thousands of genes are often found to be perturbed. Conclusion Tumor antigens are a group of proteins recognized by the cells of the immune system. Specifically, they are recognized in tumor cells where they are present in larger than usual amounts, or are physiochemically altered to a degree at which they no longer resemble native human proteins. This proteogenomic analysis of 32 tumor antigens suggests that tumor antigens have the potential to be highly specific biomarkers for different cancers.
Collapse
|
23
|
Hegde M, Moll AJ, Byrd TT, Louis CU, Ahmed N. Cellular immunotherapy for pediatric solid tumors. Cytotherapy 2014; 17:3-17. [PMID: 25082406 DOI: 10.1016/j.jcyt.2014.05.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/27/2014] [Accepted: 05/27/2014] [Indexed: 01/09/2023]
Abstract
Substantial progress has been made in the treatment of pediatric solid tumors over the past 4 decades. However, children with metastatic and or recurrent disease continue to do poorly despite the aggressive multi-modality conventional therapies. The increasing understanding of the tumor biology and the interaction between the tumor and the immune system over the recent years have led to the development of novel immune-based therapies as alternative options for some of these high-risk malignancies. The safety and anti-tumor efficacy of various tumor vaccines and tumor-antigen specific immune cells are currently being investigated for various solid tumors. In early clinical trials, most of these cellular therapies have been well tolerated and have shown promising clinical responses. Although substantial work is being done in this field, the available knowledge for pediatric tumors remains limited. We review the contemporary early phase cell-based immunotherapy efforts for pediatric solid tumors and discuss the rationale and the challenges thereof.
Collapse
Affiliation(s)
- Meenakshi Hegde
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA; Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA; Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.
| | - Alexander J Moll
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Tiara T Byrd
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA; Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Chrystal U Louis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA; Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA; Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA; Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA; Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
24
|
Abstract
Electron microscopy is a useful diagnostic technique in order to confirm or establish a definitive diagnosis in brain tumors that may have an atypical histological pattern, which requires a concrete diagnosis. In ependymomas, electron microscopy reveals morphological characters that have a pathognomonic diagnostic value, therefore allowing a definitive diagnosis. The main fine structural criteria of ependymomas consist of the numerous microvilli and cilia, which are incorporated in the cell body or extended freely in the intracellular space; the centriole or blepharoplast, which is located in the basis of the cilia; the large number of the fragmented microtubules in the perikaryon and the cellular processes (any small cellular projection into the neutrophil or intracellular space); the junctional apparatus between the cells, such as zonula adherens, zonula occludens and puncta adherentia; the basement membrane-like structure, seen in papillary ependymomas and ependymomas of the filum terminale; and the elongated cells in the loose intracellular space, commonly seen in myxopapillary ependymomas.
Collapse
Affiliation(s)
- Stavros J Baloyannis
- Aristotelian University Department of Neurology, Thessaloniki & Research Institute for Alzheimer's disease, Iraklion Langada, Greece.
| | | |
Collapse
|
25
|
Pollack IF, Jakacki RI, Butterfield LH, Okada H. Ependymomas: development of immunotherapeutic strategies. Expert Rev Neurother 2013; 13:1089-98. [PMID: 24117271 PMCID: PMC3972122 DOI: 10.1586/14737175.2013.840420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ependymomas are among the most challenging childhood brain tumors. Although 50-70% of ependymomas are cured with surgery and irradiation, a significant percentage of tumors recur. Ependymomas that are not amenable to complete resection at diagnosis have a particularly poor prognosis, and the vast majority of affected children experience tumor recurrence. Although transient responses have been observed in recurrent tumors treated with re-irradiation and several chemotherapy regimens, long-term disease control is rarely achieved. Children with recurrent disease commonly experience cumulative neurological morbidity from repeated surgical and adjuvant therapy interventions and almost universally succumb to refractory tumor progression. Accordingly, conceptually new treatment approaches are needed, both to decrease the risk of tumor recurrence and to enhance disease control in those children who experience recurrent disease. This article reviews the current application of risk-based treatment stratification at diagnosis, the rationale for exploring the role of novel therapeutic strategies such as immunotherapy at recurrence and the concept behind a vaccine-based trial for these tumors.
Collapse
Affiliation(s)
- Ian F. Pollack
- Department of Neurosurgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Regina I. Jakacki
- Department of Pediatrics, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh School of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lisa H Butterfield
- Department of Medicine, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hideho Okada
- Department of Neurosurgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|