1
|
Garcia-Moure M, Laspidea V, Gupta S, Gillard AG, Khatua S, Parthasarathy A, He J, Lang FF, Fueyo J, Alonso MM, Gomez-Manzano C. The emerging field of viroimmunotherapy for pediatric brain tumors. Neuro Oncol 2024; 26:1981-1993. [PMID: 39148489 PMCID: PMC11534321 DOI: 10.1093/neuonc/noae160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Indexed: 08/17/2024] Open
Abstract
Pediatric brain tumors are the most common solid tumors in children. Even to date, with the advances in multimodality therapeutic management, survival outcomes remain dismal in some types of tumors, such as pediatric-type diffuse high-grade gliomas or central nervous system embryonal tumors. Failure to understand the complex molecular heterogeneity and the elusive tumor and microenvironment interplay continues to undermine therapeutic efficacy. Developing a strategy that would improve survival for these fatal tumors remains unmet in pediatric neuro-oncology. Oncolytic viruses (OVs) are emerging as a feasible, safe, and promising therapy for brain tumors. The new paradigm in virotherapy implies that the direct cytopathic effect is followed, under certain circumstances, by an antitumor immune response responsible for the partial or complete debulking of the tumor mass. OVs alone or combined with other therapeutic modalities have been primarily used in adult neuro-oncology. A surge in encouraging preclinical studies in pediatric brain tumor models recently led to the clinical translation of OVs with encouraging results in these tumors. In this review, we summarize the different virotherapy tested in preclinical and clinical studies in pediatric brain tumors, and we discuss the limitations and future avenues necessary to improve the response of these tumors to this type of therapy.
Collapse
Affiliation(s)
- Marc Garcia-Moure
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Virginia Laspidea
- Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain
| | - Sumit Gupta
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew G Gillard
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Soumen Khatua
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Akhila Parthasarathy
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jiasen He
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marta M Alonso
- Department of Pediatrics, Clinica Universidad de Navarra, Pamplona, Spain
- Program of Solid Tumors, Center for the Applied Medical Research, Pamplona, Spain
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2
|
Guo Y, Li Z, Parsels LA, Wang Z, Parsels JD, Dalvi A, The S, Hu N, Valvo VM, Doherty R, Peterson E, Wang X, Venkataraman S, Agnihotri S, Venneti S, Wahl DR, Green MD, Lawrence TS, Koschmann C, Morgan MA, Zhang Q. H3K27M diffuse midline glioma is homologous recombination defective and sensitized to radiotherapy and NK cell-mediated antitumor immunity by PARP inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609803. [PMID: 39253432 PMCID: PMC11383052 DOI: 10.1101/2024.08.26.609803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Background Radiotherapy (RT) is the primary treatment for diffuse midline glioma (DMG), a lethal pediatric malignancy defined by histone H3 lysine 27-to-methionine (H3K27M) mutation. Based on the loss of H3K27 trimethylation producing broad epigenomic alterations, we hypothesized that H3K27M causes a functional double-strand break (DSB) repair defect that could be leveraged therapeutically with PARP inhibitor and RT for selective radiosensitization and antitumor immune responses. Methods H3K27M isogenic DMG cells and orthotopic brainstem DMG tumors in immune deficient and syngeneic, immune competent mice were used to evaluate the efficacy and mechanisms of PARP1/2 inhibition by olaparib or PARP1 inhibition by AZD9574 with concurrent RT. Results H3K27M mutation caused an HRR defect characterized by impaired RT-induced K63-linked polyubiquitination of histone H1 and inhibition of HRR protein recruitment. H3K27M DMG cells were selectively radiosensitized by olaparib in comparison to isogenic controls, and this effect translated to efficacy in H3K27M orthotopic brainstem tumors. Olaparib and RT induced an innate immune response and induction of NK cell (NKG2D) activating ligands leading to increased NK cell-mediated lysis of DMG tumor cells. In immunocompetent syngeneic orthotopic DMG tumors, either olaparib or AZD9574 in combination with RT enhanced intratumoral NK cell infiltration and activity in association with NK cell-mediated therapeutic responses and favorable activity of AZD9574. Conclusions The HRR deficiency in H3K27M DMG can be therapeutically leveraged with PARP inhibitors to radiosensitize and induce an NK cell-mediated antitumor immune response selectively in H3K27M DMG, supporting the clinical investigation of best-in-class PARP inhibitors with RT in DMG patients. Key points H3K27M DMG are HRR defective and selectively radiosensitized by PARP inhibitor.PARP inhibitor with RT enhances NKG2D ligand expression and NK cell-mediated lysis.NK cells are required for the therapeutic efficacy of PARP inhibitor and RT. Importance of the Study Radiotherapy is the cornerstone of H3K27M-mutant diffuse midline glioma treatment, but almost all patients succumb to tumor recurrence with poor overall survival, underscoring the need for RT-based precision combination therapy. Here, we reveal HRR deficiency as an H3K27M-mediated vulnerability and identify a novel mechanism linking impaired RT-induced histone H1 polyubiquitination and the subsequent RNF168/BRCA1/RAD51 recruitment in H3K27M DMG. This model is supported by selective radiosensitization of H3K27M DMG by PARP inhibitor. Notably, the combination treatment results in NKG2D ligand expression that confers susceptibility to NK cell killing in H3K27M DMG. We also show that the novel brain penetrant, PARP1-selective inhibitor AZD9574 compares favorably to olaparib when combined with RT, prolonging survival in a syngeneic orthotopic model of H3K27M DMG. This study highlights the ability of PARP1 inhibition to radiosensitize and induce an NK cell-mediated antitumor immunity in H3K27M DMG and supports future clinical investigation.
Collapse
|
3
|
Ausejo-Mauleon I, Labiano S, Alonso MM. Characterization of immune populations in the tumor microenvironment of diffuse midline glioma orthotopic mouse models by flow cytometry. STAR Protoc 2024; 5:102803. [PMID: 38159270 PMCID: PMC10805702 DOI: 10.1016/j.xpro.2023.102803] [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: 10/31/2023] [Revised: 11/24/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
The immune response is a fundamental process in the treatment of solid tumors. Here, we present a protocol for implanting diffuse midline glioma cells in the brain of immune-competent mice and characterizing the different immune populations in the tumor microenvironment in a flow cytometry panel. We describe steps for processing of brain tumors, isolating the immune cells, and subsequent staining with antibodies for flow cytometry. We then detail procedures for implementing the gating strategy. For complete details on the use and execution of this protocol, please refer to Ausejo-Mauleon et al.1.
Collapse
Affiliation(s)
- Iker Ausejo-Mauleon
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain; Solid Tumor Program, CIMA-Universidad de Navarra, Pamplona, Spain; Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Labiano
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain; Solid Tumor Program, CIMA-Universidad de Navarra, Pamplona, Spain; Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain.
| | - Marta M Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Spain; Solid Tumor Program, CIMA-Universidad de Navarra, Pamplona, Spain; Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
4
|
Patiño-García A, Alonso MM, Gállego Pérez-Larraya J. Promises of oncolytic viral therapy for adult and children with brain glioma. Curr Opin Oncol 2023; 35:529-535. [PMID: 37820087 DOI: 10.1097/cco.0000000000000995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to give an overview of early clinical studies addressing the safety and efficacy of oncolytic immunovirotherapy in adults and children with brain gliomas, and to highlight the extensive potential for the development of this therapeutic alternative. RECENT FINDINGS The lack of curative treatments and poor prognosis of high-grade glioma patients warrants research on innovative therapeutic alternatives such as oncolytic immunovirotherapy. Engineered modified oncolytic viruses exert both a direct lytic effect on tumor cells and a specific antitumor immune response. Early clinical trials of different DNA and RNA oncolytic viruses, mainly Herpes Simplex Virus Type-1 and adenovirus based platforms, have consistently demonstrated an acceptable safety profile, hints of efficacy and the potential of this therapy to reshape the tumor microenvironment in both adult and pediatric patients with glioma, thus constituting the basis for the development of more advanced clinical trials. SUMMARY The future landscape of oncolytic immunovirotherapy is still plenty of challenges and opportunities to enable its full therapeutic potential in both adult and children with brain gliomas.
Collapse
Affiliation(s)
- Ana Patiño-García
- Program in Solid Tumors, Center for Applied Medical Research
- Department of Pediatrics, Clínica Universidad de Navarra
- Cancer Center Clínica Universidad de Navarra
- Health Research Institute of Navarra (IdiSNA)
| | - Marta M Alonso
- Program in Solid Tumors, Center for Applied Medical Research
- Department of Pediatrics, Clínica Universidad de Navarra
- Cancer Center Clínica Universidad de Navarra
- Health Research Institute of Navarra (IdiSNA)
| | - Jaime Gállego Pérez-Larraya
- Program in Solid Tumors, Center for Applied Medical Research
- Cancer Center Clínica Universidad de Navarra
- Health Research Institute of Navarra (IdiSNA)
- Department of Neurology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| |
Collapse
|
5
|
Gállego Pérez-Larraya J, García-Moure M, Alonso MM. Oncolytic virotherapy for the treatment of pediatric brainstem gliomas. Rev Neurol (Paris) 2023; 179:475-480. [PMID: 37061388 DOI: 10.1016/j.neurol.2023.03.016] [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: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/17/2023]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is the most frequent brainstem glioma and the most lethal brain tumor in childhood. Despite transient benefit with radiotherapy, the prognosis of children with this disease remains dismal with severe neurological morbidity and median survival less than 12months. Oncolytic immunovirotherapy is emerging as a potential therapeutic approach in neuro-oncology. The oncolytic adenovirus Delta-24-RGD has shown efficacy in adult patients with recurrent GBM. Our group has demonstrated that Delta-24-RGD has oncolytic activity and triggers immune response in preclinical models of DIPG, and has a synergistic effect with radiotherapy in animal models of this disease. In this scenario, we conducted a first-in-human phase 1 clinical trial to evaluate the safety and efficacy of intratumoral injection of Delta-24-RGD in pediatric patients with newly diagnosed DIPG prior to standard radiotherapy. The study confirmed the feasibility of this treatment with an acceptable safety profile and encouraging efficacy results. Correlative analyses showed a biological activity from Delta-24-RGD in DIPG. Further advanced trials are needed to validate these results. Meanwhile, plenty of opportunities to increase the potential contribution of oncolytic viruses in the management of devastating tumors with no current effective treatment such as DIPG need to be explored and exploited.
Collapse
Affiliation(s)
- Jaime Gállego Pérez-Larraya
- Program in Solid Tumors, Center for Applied Medical Research, Pamplona, Navarra, Spain; Department of Neurology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain.
| | - Marc García-Moure
- Program in Solid Tumors, Center for Applied Medical Research, Pamplona, Navarra, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
| | - Marta M Alonso
- Program in Solid Tumors, Center for Applied Medical Research, Pamplona, Navarra, Spain; Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Navarra, Spain; Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
| |
Collapse
|
6
|
Jovanovich N, Habib A, Head J, Hameed F, Agnihotri S, Zinn PO. Pediatric diffuse midline glioma: Understanding the mechanisms and assessing the next generation of personalized therapeutics. Neurooncol Adv 2023; 5:vdad040. [PMID: 37152806 PMCID: PMC10162114 DOI: 10.1093/noajnl/vdad040] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Diffuse midline glioma (DMG) is a pediatric cancer that originates in the midline structures of the brain. Prognosis of DMG patients remains poor due to the infiltrative nature of these tumors and the protection they receive from systemically delivered therapeutics via an intact blood-brain barrier (BBB), making treatment difficult. While the cell of origin remains disputed, it is believed to reside in the ventral pons. Recent research has pointed toward epigenetic dysregulation inducing an OPC-like transcriptomic signature in DMG cells. This epigenetic dysregulation is typically caused by a mutation (K27M) in one of two histone genes-H3F3A or HIST1H3B -and can lead to a differentiation block that increases these cells oncogenic potential. Standard treatment with radiation is not sufficient at overcoming the aggressivity of this cancer and only confers a survival benefit of a few months, and thus, discovery of new therapeutics is of utmost importance. In this review, we discuss the cell of origin of DMGs, as well as the underlying molecular mechanisms that contribute to their aggressivity and resistance to treatment. Additionally, we outline the current standard of care for DMG patients and the potential future therapeutics for this cancer that are currently being tested in preclinical and clinical trials.
Collapse
Affiliation(s)
- Nicolina Jovanovich
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jeffery Head
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Farrukh Hameed
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sameer Agnihotri
- Sameer Agnihtroi, PhD, 4401 Penn Avenue, Office 7126, Pittsburgh, PA 15224, USA ()
| | - Pascal O Zinn
- Corresponding Authors: Pascal O. Zinn, MD, PhD, 5150 Centre Ave. Suite 433, Pittsburgh, PA 15232, USA ()
| |
Collapse
|
7
|
Laspidea V, Puigdelloses M, Labiano S, Marrodán L, Garcia-Moure M, Zalacain M, Gonzalez-Huarriz M, Martínez-Vélez N, Ausejo-Mauleon I, de la Nava D, Herrador-Cañete G, Marco-Sanz J, Guruceaga E, de Andrea CE, Villalba M, Becher O, Squatrito M, Matía V, Gállego Pérez-Larraya J, Patiño-García A, Gupta S, Gomez-Manzano C, Fueyo J, Alonso MM. Exploiting 4-1BB immune checkpoint to enhance the efficacy of oncolytic virotherapy for diffuse intrinsic pontine gliomas. JCI Insight 2022; 7:154812. [PMID: 35393952 PMCID: PMC9057625 DOI: 10.1172/jci.insight.154812] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/25/2022] [Indexed: 12/28/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors, and patient survival has not changed despite many therapeutic efforts, emphasizing the urgent need for effective treatments. Here, we evaluated the anti-DIPG effect of the oncolytic adenovirus Delta-24-ACT, which was engineered to express the costimulatory ligand 4-1BBL to potentiate the antitumor immune response of the virus. Delta-24-ACT induced the expression of functional 4-1BBL on the membranes of infected DIPG cells, which enhanced the costimulation of CD8+ T lymphocytes. In vivo, Delta-24-ACT treatment of murine DIPG orthotopic tumors significantly improved the survival of treated mice, leading to long-term survivors that developed immunological memory against these tumors. In addition, Delta-24-ACT was safe and caused no local or systemic toxicity. Mechanistic studies showed that Delta-24-ACT modulated the tumor-immune content, not only increasing the number, but also improving the functionality of immune cells. All of these data highlight the safety and potential therapeutic benefit of Delta-24-ACT the treatment of patients with DIPG.
Collapse
Affiliation(s)
- Virginia Laspidea
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Montserrat Puigdelloses
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Sara Labiano
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Lucía Marrodán
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Marc Garcia-Moure
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Marta Zalacain
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Marisol Gonzalez-Huarriz
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Naiara Martínez-Vélez
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Iker Ausejo-Mauleon
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Daniel de la Nava
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Guillermo Herrador-Cañete
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Gene Therapy and Regulation of Gene Expression Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
| | - Javier Marco-Sanz
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Elisabeth Guruceaga
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Bioinformatics Platform, El Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Carlos E de Andrea
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Department of Pathology, Navarra University Clinic, Pamplona, Spain
| | - María Villalba
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Department of Pathology, Navarra University Clinic, Pamplona, Spain
| | - Oren Becher
- Department of Pediatrics.,Department of Biochemistry and Molecular Genetics, and.,Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA.,Division of Hematology Oncology and Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital, Chicago, Illinois, USA
| | - Massimo Squatrito
- Seve Ballesteros Foundation Brain Tumor Group, Molecular Oncology Programme, Spanish National Cancer Research Center, Madrid, Spain
| | - Verónica Matía
- Seve Ballesteros Foundation Brain Tumor Group, Molecular Oncology Programme, Spanish National Cancer Research Center, Madrid, Spain
| | - Jaime Gállego Pérez-Larraya
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Neurology, Navarra University Clinic, Pamplona, Spain
| | - Ana Patiño-García
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| | - Sumit Gupta
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marta M Alonso
- Health Research Institute of Navarra, Pamplona, Navarra, Spain.,Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain.,Department of Pediatrics, Navarra University Clinic, Pamplona, Spain
| |
Collapse
|
8
|
Faisal SM, Mendez FM, Nunez F, Castro MG, Lowenstein PR. Immune-stimulatory (TK/Flt3L) gene therapy opens the door to a promising new treatment strategy against brainstem gliomas. Oncotarget 2020; 11:4607-4612. [PMID: 33400737 PMCID: PMC7747859 DOI: 10.18632/oncotarget.27834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a rare brainstem tumor which carries a dismal prognosis. To date. there are no effective treatments for DIPG. Transcriptomic studies have shown that DIPGs have a distinct profile compared to hemispheric high-grade pediatric gliomas. These specific genomic features coupled with the younger median age group suggest that DIPG is of developmental origin. There is a major unmet need for novel effective therapeutic approaches for DIPG. Clinical and preclinical studies have expanded our understanding of the molecular pathways in this deadly disease. We have developed a genetically engineered brainstem glioma model harboring the recurrent DIPG mutation, activin A receptor type I (ACVR1)-G328V (mACVR1) using the sleeping beauty transposon system. DIPG neurospheres isolated from the genetically engineered mouse model were implanted into the pons of immune-competent mice to assess the therapeutic efficacy and toxicity of immunostimulatory gene therapy using adenoviruses expressing thymidine kinase (TK) and fms-like tyrosine kinase 3 ligand (Flt3L). Immunostimulatory adenoviral-mediated delivery of TK/Flt3L in mice bearing brainstem gliomas resulted in antitumor immunity, recruitment of antitumor-specific T cells, and improved median survival by stimulating the host antitumor immune response. Therapeutic efficacy of the immunostimulatory gene therapy strategy will be tested in the clinical arena in a Phase I clinical trial. We also discuss immunotherapeutic interventions currently being implemented in DIPG patients and discuss the profound therapeutic implications of immunotherapy for this patient populations.
Collapse
Affiliation(s)
- Syed M. Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Flor M. Mendez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fernando Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
The oncolytic virus Delta-24-RGD elicits an antitumor effect in pediatric glioma and DIPG mouse models. Nat Commun 2019; 10:2235. [PMID: 31138805 PMCID: PMC6538754 DOI: 10.1038/s41467-019-10043-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 04/16/2019] [Indexed: 12/17/2022] Open
Abstract
Pediatric high-grade glioma (pHGG) and diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors in desperate need of a curative treatment. Oncolytic virotherapy is emerging as a solid therapeutic approach. Delta-24-RGD is a replication competent adenovirus engineered to replicate in tumor cells with an aberrant RB pathway. This virus has proven to be safe and effective in adult gliomas. Here we report that the administration of Delta-24-RGD is safe in mice and results in a significant increase in survival in immunodeficient and immunocompetent models of pHGG and DIPGs. Our results show that the Delta-24-RGD antiglioma effect is mediated by the oncolytic effect and the immune response elicited against the tumor. Altogether, our data highlight the potential of this virus as treatment for patients with these tumors. Of clinical significance, these data have led to the start of a phase I/II clinical trial at our institution for newly diagnosed DIPG (NCT03178032).
Collapse
|
10
|
Welby JP, Kaptzan T, Wohl A, Peterson TE, Raghunathan A, Brown DA, Gupta SK, Zhang L, Daniels DJ. Current Murine Models and New Developments in H3K27M Diffuse Midline Gliomas. Front Oncol 2019; 9:92. [PMID: 30873381 PMCID: PMC6400847 DOI: 10.3389/fonc.2019.00092] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/31/2019] [Indexed: 01/24/2023] Open
Abstract
Diffuse Midline Gliomas with Histone 3-Lysine-27-Methionine (H3K27M) mutation constitute the majority of Diffuse Intrinsic Pontine Glioma (DIPG), which is the most aggressive form of pediatric glioma with a dire prognosis. DIPG are lethal tumors found in younger children with a median survival <1 year from diagnosis. Discovery of the characteristic H3K27M mutations offers opportunity and hope for development of targeted therapies for this deadly disease. The H3K27M mutation, likely through epigenetic alterations in specific H3 lysine trimethylation levels and subsequent gene expression, plays a significant role in pathogenesis of DIPG. Animal models accurately depicting molecular characteristics of H3K27M DIPG are important to elucidate underlying pathologic events and for preclinical drug evaluation. Here we review the past and present DIPG models and describe our efforts developing patient derived cell lines and xenografts from pretreated surgical specimens. Pre-treated surgical samples retain the characteristic genomic and phenotypic hallmarks of DIPG and establish orthotopic tumors in the mouse brainstem that recapitulate radiographic and morphological features of the original human DIPG tumor. These models that contain the H3K27M mutation constitute a valuable tool to further study this devastating disease and ultimately may uncover novel therapeutic vulnerabilities.
Collapse
Affiliation(s)
- John P Welby
- Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tatiana Kaptzan
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Anton Wohl
- Department of Neurosurgery, Chaim Sheba Medical Center, Tel-HaShomer, Ramat-Gan, Israel
| | | | - Aditya Raghunathan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Desmond A Brown
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States
| | - Shiv K Gupta
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| | - Liang Zhang
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States
| | - David J Daniels
- Department of Neurosurgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
11
|
Mathew RK, Rutka JT. Diffuse Intrinsic Pontine Glioma : Clinical Features, Molecular Genetics, and Novel Targeted Therapeutics. J Korean Neurosurg Soc 2018; 61:343-351. [PMID: 29742880 PMCID: PMC5957322 DOI: 10.3340/jkns.2018.0008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 01/21/2018] [Indexed: 12/18/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a deadly paediatric brain cancer. Transient response to radiation, ineffective chemotherapeutic agents and aggressive biology result in rapid progression of symptoms and a dismal prognosis. Increased availability of tumour tissue has enabled the identification of histone gene aberrations, genetic driver mutations and methylation changes, which have resulted in molecular and phenotypic subgrouping. However, many of the underlying mechanisms of DIPG oncogenesis remain unexplained. It is hoped that more representative in vitro and preclinical models–using both xenografted material and genetically engineered mice–will enable the development of novel chemotherapeutic agents and strategies for targeted drug delivery. This review provides a clinical overview of DIPG, the barriers to progress in developing effective treatment, updates on drug development and preclinical models, and an introduction to new technologies aimed at enhancing drug delivery.
Collapse
Affiliation(s)
- Ryan K Mathew
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK.,Department of Neurosurgery, Leeds General Infirmary, Leeds, UK
| | - James T Rutka
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
12
|
Tejada S, Díez-Valle R, Domínguez PD, Patiño-García A, González-Huarriz M, Fueyo J, Gomez-Manzano C, Idoate MA, Peterkin J, Alonso MM. DNX-2401, an Oncolytic Virus, for the Treatment of Newly Diagnosed Diffuse Intrinsic Pontine Gliomas: A Case Report. Front Oncol 2018; 8:61. [PMID: 29594041 PMCID: PMC5858123 DOI: 10.3389/fonc.2018.00061] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/22/2018] [Indexed: 12/15/2022] Open
Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are aggressive glial brain tumors that primarily affect children, for which there is no curative treatment. Median overall survival is only one year. Currently, the scientific focus is on expanding the knowledge base of the molecular biology of DIPG, and identifying effective therapies. Oncolytic adenovirus DNX-2401 is a replication-competent, genetically modified virus capable of infecting and killing glioma cells, and stimulating an anti-tumor immune response. Clinical trials evaluating intratumoral DNX-2401 in adults with recurrent glioblastoma have demonstrated that the virus has a favorable safety profile and can prolong survival. Subsequently, these results have encouraged the transition of this biologically active therapy from adults into the pediatric population. To this aim, we have designed a clinical Phase I trial for newly diagnosed pediatric DIPG to investigate the feasibility, safety, and preliminary efficacy of delivering DNX-2401 into tumors within the pons following biopsy. This case report presents a pediatric patient enrolled in this ongoing Phase I trial for children and adolescents with newly diagnosed DIPG. The case involves an 8-year-old female patient with radiologically diagnosed DIPG who underwent stereotactic tumor biopsy immediately followed by intratumoral DNX-2401 in the same biopsy track. Because there were no safety concerns or new neurological deficits, the patient was discharged 3 days after the procedures. To our knowledge, this is the first report of intratumoral DNX-2401 for a patient with DIPG in a clinical trial. We plan to demonstrate that intratumoral delivery of an oncolytic virus following tumor biopsy for pediatric patients with DIPG is a novel and feasible approach and that DNX-2401 represents an innovative treatment for the disease.
Collapse
Affiliation(s)
- Sonia Tejada
- Department of Neurosurgery, University Hospital of Navarra, Pamplona, Spain.,The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
| | - Ricardo Díez-Valle
- Department of Neurosurgery, University Hospital of Navarra, Pamplona, Spain.,The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
| | - Pablo D Domínguez
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Radiology, University Hospital of Navarra, Pamplona, Spain
| | - Ana Patiño-García
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| | - Marisol González-Huarriz
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain
| | - Juan Fueyo
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cande Gomez-Manzano
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | - Marta M Alonso
- The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.,Program in Solid Tumors and Biomarkers, Foundation for the Applied Medical Research, Pamplona, Spain.,Department of Pediatrics, University Hospital of Navarra, Pamplona, Spain
| |
Collapse
|