1
|
Guo Q, Yang W, Robinson G, Chaludiya K, Abdulkadir AN, Roy FG, Shivakumar D, Ahmad AN, Abdulkadir SA, Kirschner AN. Unlocking the Radiosensitizing Potential of MYC inhibition in Neuroendocrine Malignancies. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00431-6. [PMID: 40354951 DOI: 10.1016/j.ijrobp.2025.04.034] [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: 11/03/2024] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
The MYC family of transcription factors-comprising c-MYC, N-MYC, and L-MYC-plays a pivotal role in oncogenesis, driving cancer progression and resistance to therapy. While MYC proteins have long been considered challenging drug targets due to their intricate structures, recent advances have led to the development of promising inhibitors. This review explores the role of MYC overexpression in promoting radiotherapy resistance in aggressive neuroendocrine malignancies through multiple mechanisms, including increased tumor cell invasion, enhanced DNA damage repair and oxidative stress management, pro-survival autophagy, survival of circulating tumor cells, angiogenesis, awakening from dormancy, and modulation of chronic inflammation and host immunity. Paradoxically, MYC overexpression can also enhance radiosensitivity in certain cancer cells by driving pro-apoptotic pathways, such as reactive oxygen species (ROS)-induced DNA damage that overwhelms cellular repair mechanisms, ultimately leading to cell death. Additionally, we provide a comprehensive summary of direct MYC inhibitors, detailing their current stage of preclinical and clinical development as novel anti-cancer therapeutics. This review highlights MYC's role in cancer metastasis and radiotherapy resistance while examining the potential of MYC inhibitors as radiosensitizers in adult and pediatric neuroendocrine malignancies, including small cell lung cancer, large cell neuroendocrine lung cancer, Merkel cell carcinoma, neuroendocrine-differentiated prostate cancer, neuroblastoma, central nervous system embryonal tumors, and medulloblastoma.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA 32224; Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA 32224; Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA; Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611.
| | - William Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611.
| | - Guy Robinson
- Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA 32224; Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| | - Keyur Chaludiya
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905
| | | | - Falguni Ghosh Roy
- MGM Institute of Health Sciences, Navi Mumbai, Maharashtra, India 410209
| | - Divya Shivakumar
- Kamineni Academy of Medical Science and Research Centre, Hyderabad, Telangana, India 500068
| | - Ayesha N Ahmad
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; Boonshoft School of Medicine, Wright State University, Fairborn, OH, USA 45324
| | - Sarki A Abdulkadir
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611; The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA 60611.
| | - Austin N Kirschner
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA 37232.
| |
Collapse
|
2
|
Watson PM, DeVaux CA, Freeman KW. Loss of ARID1A leads to a cold tumor phenotype via suppression of IFNγ signaling. Sci Rep 2025; 15:8716. [PMID: 40082458 PMCID: PMC11906763 DOI: 10.1038/s41598-025-91688-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/21/2025] [Indexed: 03/16/2025] Open
Abstract
The collapse of inflammatory signaling that recruits cytotoxic immune cells to the tumor microenvironment contributes to the immunologically cold tumor phenotype in neuroblastoma (NB) and is a barrier to NB immunotherapy. Multiple studies have reported that MYCN amplification, a trait of high-risk NB, correlates with a loss of inflammatory signaling; but MYCN also correlates with 1p36 deletions in NB where the SWI/SNF chromatin remodeling complex subunit ARID1A (1p36.11) is located. ARID1A is known to support inflammatory signaling in adult cancers but its role in NB inflammatory signaling is unexplored. We find MYCN overexpression causes a stronger inflammatory response to interferon-gamma (IFNγ). ARID1A knockdown causes a weaker inflammatory response and reduces IFNγ induced gene signatures for the transcription factor interferon response factor 1 (IRF1). We found ARID1A is a functional interactor of IRF1 by co-immunoprecipitation studies, and ARID1A silencing causes loss of activating chromatin marks at the IRF1 target gene CXCL10. We model that IRF1 uses ARID1A containing SWI/SNF to promote CXCL10 in response to IFNγ. Our work clarifies that the loss of ARID1A, which tightly associates with MYCN amplification, causes reduced inflammatory signaling. This work finds that ARID1A is a critical regulator of inflammatory signaling in NB and provides rationale for testing immune therapies in MYCN amplified NB that are effective in adult ARID1A mutated cancers.
Collapse
Affiliation(s)
- Pamela M Watson
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Chelsea A DeVaux
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kevin W Freeman
- Department of Genetics, Genomics, and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA.
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
3
|
Wang W, Du Y, Datta S, Fowler JF, Sang HT, Albadari N, Li W, Foster J, Zhang R. Targeting the MYCN-MDM2 pathways for cancer therapy: Are they druggable? Genes Dis 2025; 12:101156. [PMID: 39802403 PMCID: PMC11719324 DOI: 10.1016/j.gendis.2023.101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/11/2023] [Accepted: 09/26/2023] [Indexed: 01/16/2025] Open
Abstract
Targeting oncogenes and their interactive partners is an effective approach to developing novel targeted therapies for cancer and other chronic diseases. We and others have long suggested the MDM2 oncogene being an excellent target for cancer therapy, based on its p53-dependent and -independent oncogenic activities in a variety of cancers. The MYC family proteins are transcription factors that also regulate diverse biological functions. Dysregulation of MYC, such as amplification of MYCN, is associated with tumorigenesis, especially for neuroblastoma. Although the general survival rate of neuroblastoma patients has significantly improved over the past few decades, high-risk neuroblastoma still presents a poor prognosis. Therefore, innovative and more potent therapeutic strategies are needed to eradicate these aggressive neoplasms. This review focuses on the oncogenic properties of MYCN and its molecular regulation and summarizes the major therapeutic strategies being developed based on preclinical findings. We also highlight the potential benefits of targeting both the MYCN and MDM2 oncogenes, providing preclinical evidence of the efficacy and safety of this approach. In conclusion, the development of effective small molecules that inhibit both MYCN and MDM2 represents a promising new strategy for the treatment of neuroblastoma and other cancers.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
- Drug Discovery Institute, University of Houston, Houston, TX 77204, USA
| | - Yi Du
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Josef F. Fowler
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Hannah T. Sang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Najah Albadari
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jennifer Foster
- Texas Children's Hospital, Department of Pediatrics, Section of Hematology-Oncology Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
- Drug Discovery Institute, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
4
|
Bley IA, Behrens S, Spohn M, Müller I, Schattling B. Genetic Risk Profiling Reveals Altered Glycosyltransferase Expression as a Predictor for Patient Outcome in Neuroblastoma. J Clin Med 2025; 14:527. [PMID: 39860532 PMCID: PMC11766279 DOI: 10.3390/jcm14020527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/17/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Neuroblastoma is a highly aggressive pediatric cancer that arises from immature nerve cells and exhibits a broad spectrum of clinical presentations. While low- and intermediate-risk neuroblastomas often have favorable outcomes, high-risk neuroblastomas are associated with poor prognosis and significant treatment challenges. The complex genetic networks driving these high-risk cases remain poorly understood. This study aims to investigate differences in gene expression patterns that may contribute to disease outcomes. Methods: We employed an in silico approach to analyze a cohort of 493 neuroblastoma tumor samples that underwent mRNA sequencing (GSE49711). This dataset was reanalyzed in depth with a non-hypothesis-driven approach to identify the expression patterns and regulatory mechanisms associated with a poor prognosis. Results: By exploring global gene expression and the integration of clinical parameters, we stratified the samples into two groups with highly distinct gene expression profiles. MYCN amplification emerged as a major driver not only of poor prognosis but also of specific gene regulatory patterns. Notably, tumors with MYCN amplification exhibited the strong regulation of immune response genes and less immune infiltration, suggesting potential immune evasion. However, while we observed only minor changes in immune checkpoint expression, there was a strong modulation of glycosyltransferase genes in MYCN-amplified tumors. Using this information, we were able to construct a risk profile based on 12 glycosylation-related genes, which correlates with the survival outcomes of neuroblastoma patients. Conclusions: This study highlights the role of MYCN amplification in driving a poor prognosis in neuroblastoma through the regulation of immune response and glycosylation-related genes. Based on this finding, we developed a genetic risk profile that correlates with survival outcomes in neuroblastoma patients.
Collapse
Affiliation(s)
- Isabelle Ariane Bley
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Behrens
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
| | - Michael Spohn
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ingo Müller
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Benjamin Schattling
- Research Institute Children’s Cancer Center Hamburg, 20251 Hamburg, Germany
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
5
|
Wertheim KY, Chisholm R, Richmond P, Walker D. Multicellular model of neuroblastoma proposes unconventional therapy based on multiple roles of p53. PLoS Comput Biol 2024; 20:e1012648. [PMID: 39715281 PMCID: PMC11723635 DOI: 10.1371/journal.pcbi.1012648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 01/10/2025] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Neuroblastoma is the most common extra-cranial solid tumour in children. Over half of all high-risk cases are expected to succumb to the disease even after chemotherapy, surgery, and immunotherapy. Although the importance of MYCN amplification in this disease is indisputable, the mechanistic details remain enigmatic. Here, we present a multicellular model of neuroblastoma comprising a continuous automaton, discrete cell agents, and a centre-based mechanical model, as well as the simulation results we obtained with it. The continuous automaton represents the tumour microenvironment as a grid-like structure, where each voxel is associated with continuous variables such as the oxygen level therein. Each discrete cell agent is defined by several attributes, including its cell cycle position, mutations, gene expression pattern, and more with behaviours such as cell cycling and cell death being stochastically dependent on these attributes. The centre-based mechanical model represents the properties of these agents as physical objects, describing how they repel each other as soft spheres. By implementing a stochastic simulation algorithm on modern GPUs, we simulated the dynamics of over one million neuroblastoma cells over a period of months. Specifically, we set up 1200 heterogeneous tumours and tracked the MYCN-amplified clone's dynamics in each, revealed the conditions that favour its growth, and tested its responses to 5000 drug combinations. Our results are in agreement with those reported in the literature and add new insights into how the MYCN-amplified clone's reproductive advantage in a tumour, its gene expression profile, the tumour's other clones (with different mutations), and the tumour's microenvironment are inter-related. Based on the results, we formulated a hypothesis, which argues that there are two distinct populations of neuroblastoma cells in the tumour; the p53 protein is pro-survival in one and pro-apoptosis in the other. It follows that alternating between inhibiting MDM2 to restore p53 activity and inhibiting ARF to attenuate p53 activity is a promising, if unorthodox, therapeutic strategy. The multicellular model has the advantages of modularity, high resolution, and scalability, making it a potential foundation for creating digital twins of neuroblastoma patients.
Collapse
Affiliation(s)
- Kenneth Y. Wertheim
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
- Centre of Excellence for Data Science, Artificial Intelligence, and Modelling, University of Hull, Kingston upon Hull, United Kingdom
- School of Computer Science, University of Hull, Kingston upon Hull, United Kingdom
| | - Robert Chisholm
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Paul Richmond
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
| | - Dawn Walker
- Insigneo Institute for in Silico Medicine, University of Sheffield, Sheffield, United Kingdom
- School of Computer Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
6
|
Maher S, Wynne K, Zhernovkov V, Halasz M. A temporal (phospho-)proteomic dataset of neurotrophic receptor tyrosine kinase signalling in neuroblastoma. Sci Data 2024; 11:1111. [PMID: 39389992 PMCID: PMC11467210 DOI: 10.1038/s41597-024-03965-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024] Open
Abstract
Neurotrophic receptor tyrosine kinases (TrkA, TrkB, TrkC), despite their homology, contribute to the clinical heterogeneity of the childhood cancer neuroblastoma. TrkA expression is associated with low-stage disease and is often seen with spontaneous tumour regression. Conversely, TrkB is present in unfavourable neuroblastomas that often harbour amplification of the MYCN oncogene. The role of TrkC is less clearly defined, although some studies suggest its association with a favourable outcome. Understanding the differences in activity of Trk receptors that drive divergent clinical phenotypes as well as the influence of MYCN amplification on downstream Trk receptor signalling remains poorly understood. Here, we present a comprehensive label-free mass spectrometry-based total proteomics and phosphoproteomics dataset (432 raw files with FragPipe search outputs; available on PRIDE with accession number PXD054441) where we identified and quantified 4,907 proteins, 16,744 phosphosites and 5,084 phosphoproteins, derived from NGF/BDNF/NT-3 treated TrkA/B/C-overexpressing neuroblastoma cells with differential MYCN status. Analysing our dataset offers valuable insights into TrkA/B/C receptor signalling in neuroblastoma and its modulation by MYCN status; and holds potential for advancing therapeutic strategies in this challenging childhood cancer.
Collapse
Affiliation(s)
- Stephanie Maher
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Kieran Wynne
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Vadim Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Melinda Halasz
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
7
|
Kaufman ME, Vayani OR, Moore K, Chlenski A, Wu T, Lee SM, Desai AV, He C, Cohn SL, Applebaum MA. Characterizing Relationships between T-cell Inflammation and Outcomes in Patients with High-Risk Neuroblastoma According to Mesenchymal and Adrenergic Signatures. CANCER RESEARCH COMMUNICATIONS 2024; 4:2255-2266. [PMID: 39099200 PMCID: PMC11350481 DOI: 10.1158/2767-9764.crc-24-0214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 07/12/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
Recent insights have identified adrenergic (ADRN) and mesenchymal (MES) cell lineages as distinct biologic cell types and T-cell inflammation as a prognostic marker in neuroblastoma. We hypothesized that elucidating unique and overlapping aspects of these biologic features could serve as novel biomarkers for informing ongoing efforts to improve therapeutic approaches for children with high-risk neuroblastoma. We identified lineage-specific, single-stranded super-enhancers to define ADRN and MES specific genes. Publicly available RNA-seq of diagnostic tumor biopsies was used in Discovery and Validation cohorts. Each tumor was assigned a relative MES score and T-cell inflammation (TCI) score. Survival was assessed using the Kaplan-Meier method, and differences were assessed by the log-rank test. Inflammation scores were correlated with MES scores and anticorrelated with MYCN-amplification in both cohorts. Among patients with high-risk, ADRN tumors, those with TCI tumors had superior overall survival to those with non-inflamed tumors. A similar, but nonsignificant, trend was observed in the Validation cohort. Conversely, there was no difference according to TCI status in the MES cohort in either the Discover or Validation cohorts. High-inflammation scores were correlated with improved survival in some patients with high-risk, ADRN but not MES neuroblastoma. Our findings bolster support for further developing T-cell-based and immunotherapy-based approaches for children with high-risk neuroblastoma of varying MES and ADRN expression. SIGNIFICANCE Adrenergic (ADRN) and mesenchymal (MES) lineages are distinct biologic cell types in neuroblastoma. We defined ADRN and MES specific genes and found that high-risk, ADRN tumors harboring elevated T-cell inflammation signatures had superior overall survival. Our findings bolster support for further developing immunotherapy-based approaches for children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Maria E. Kaufman
- The University of Chicago Pritzker School of Medicine, Chicago, Illinois.
| | - Omar R. Vayani
- The University of Chicago Pritzker School of Medicine, Chicago, Illinois.
| | - Kelley Moore
- Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois.
| | - Alexandre Chlenski
- Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois.
| | - Tong Wu
- Department of Chemistry, University of Chicago, Chicago, Illinois.
| | - Sang Mee Lee
- Biostatistics Laboratory and Research Computing Group, The University of Chicago, Chicago, Illinois.
| | - Ami V. Desai
- Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois.
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, Illinois.
- Howard Hughes Medical Institute, Chevy Chase. Maryland.
| | - Susan L. Cohn
- Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois.
| | - Mark A. Applebaum
- Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
8
|
Chu Y, Nayyar G, Tian M, Lee DA, Ozkaynak MF, Ayala-Cuesta J, Klose K, Foley K, Mendelowitz AS, Luo W, Liao Y, Ayello J, Behbehani GK, Riddell S, Cripe T, Cairo MS. Efficiently targeting neuroblastoma with the combination of anti-ROR1 CAR NK cells and N-803 in vitro and in vivo in NB xenografts. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200820. [PMID: 38933492 PMCID: PMC11201149 DOI: 10.1016/j.omton.2024.200820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
The prognosis for children with recurrent and/or refractory neuroblastoma (NB) is dismal. The receptor tyrosine kinase-like orphan receptor 1 (ROR1), which is highly expressed on the surface of NB cells, provides a potential target for novel immunotherapeutics. Anti-ROR1 chimeric antigen receptor engineered ex vivo expanded peripheral blood natural killer (anti-ROR1 CAR exPBNK) cells represent this approach. N-803 is an IL-15 superagonist with enhanced biological activity. In this study, we investigated the in vitro and in vivo anti-tumor effects of anti-ROR1 CAR exPBNK cells with or without N-803 against ROR1+ NB models. Compared to mock exPBNK cells, anti-ROR1 CAR exPBNK cells had significantly enhanced cytotoxicity against ROR1+ NB cells, and N-803 further increased cytotoxicity. High-dimensional analysis revealed that N-803 enhanced Stat5 phosphorylation and Ki67 levels in both exPBNK and anti-ROR1 CAR exPBNK cells with or without NB cells. In vivo, anti-ROR1 CAR exPBNK plus N-803 significantly (p < 0.05) enhanced survival in human ROR1+ NB xenografted NSG mice compared to anti-ROR1 CAR exPBNK alone. Our results provide the rationale for further development of anti-ROR1 CAR exPBNK cells plus N-803 as a novel combination immunotherapeutic for patients with recurrent and/or refractory ROR1+ NB.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Gaurav Nayyar
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Meijuan Tian
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Dean A. Lee
- Department of Pediatric Hem/Onc/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Mehmet F. Ozkaynak
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | | | - Kayleigh Klose
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Keira Foley
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | | | - Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Yanling Liao
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
| | - Gregory K. Behbehani
- Department of Internal Medicine, Division of Hematology, the Ohio State University; Columbus, OH 43210, USA
| | - Stanley Riddell
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Tim Cripe
- Department of Pediatric Hem/Onc/BMT, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY 10595, USA
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA
- Department of Microbiology, Immunology and Pathology, New York Medical College, Valhalla, NY 10595, USA
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
9
|
Miranda A, Pattnaik S, Hamilton PT, Fuss MA, Kalaria S, Laumont CM, Smazynski J, Mesa M, Banville A, Jiang X, Jenkins R, Cañadas I, Nelson BH. N-MYC impairs innate immune signaling in high-grade serous ovarian carcinoma. SCIENCE ADVANCES 2024; 10:eadj5428. [PMID: 38748789 PMCID: PMC11095474 DOI: 10.1126/sciadv.adj5428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is a challenging disease, especially for patients with immunologically "cold" tumors devoid of tumor-infiltrating lymphocytes (TILs). We found that HGSC exhibits among the highest levels of MYCN expression and transcriptional signature across human cancers, which is strongly linked to diminished features of antitumor immunity. N-MYC repressed basal and induced IFN type I signaling in HGSC cell lines, leading to decreased chemokine expression and T cell chemoattraction. N-MYC inhibited the induction of IFN type I by suppressing tumor cell-intrinsic STING signaling via reduced STING oligomerization, and by blunting RIG-I-like receptor signaling through inhibition of MAVS aggregation and localization in the mitochondria. Single-cell RNA sequencing of human clinical HGSC samples revealed a strong negative association between cancer cell-intrinsic MYCN transcriptional program and type I IFN signaling. Thus, N-MYC inhibits tumor cell-intrinsic innate immune signaling in HGSC, making it a compelling target for immunotherapy of cold tumors.
Collapse
Affiliation(s)
- Alex Miranda
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Swetansu Pattnaik
- The Kinghorn Cancer Centre and Cancer Division, Garvan Institute of Medical Research, 370 Victoria St, Darlinghurst, NSW, Australia
| | - Phineas T. Hamilton
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Shreena Kalaria
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
| | - Céline M. Laumont
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | | | - Monica Mesa
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 3E6, Canada
| | - Allyson Banville
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Xinpei Jiang
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Russell Jenkins
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Israel Cañadas
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Brad H. Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 3E6, Canada
| |
Collapse
|
10
|
Maines LW, Keller SN, Smith RA, Schrecengost RS, Smith CD. Opaganib Downregulates N-Myc Expression and Suppresses In Vitro and In Vivo Growth of Neuroblastoma Cells. Cancers (Basel) 2024; 16:1779. [PMID: 38730731 PMCID: PMC11082966 DOI: 10.3390/cancers16091779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroblastoma (NB), the most common cancer in infants and the most common solid tumor outside the brain in children, grows aggressively and responds poorly to current therapies. We have identified a new drug (opaganib, also known as ABC294640) that modulates sphingolipid metabolism by inhibiting the synthesis of sphingosine 1-phosphate (S1P) by sphingosine kinase-2 and elevating dihydroceramides by inhibition of dihydroceramide desaturase. The present studies sought to determine the potential therapeutic activity of opaganib in cell culture and xenograft models of NB. Cytotoxicity assays demonstrated that NB cells, including cells with amplified MYCN, are effectively killed by opaganib concentrations well below those that accumulate in tumors in vivo. Opaganib was shown to cause dose-dependent decreases in S1P and hexosylceramide levels in Neuro-2a cells, while concurrently elevating levels of dihydroceramides. As with other tumor cells, opaganib reduced c-Myc and Mcl-1 protein levels in Neuro-2a cells, and also reduced the expression of the N-Myc protein. The in vivo growth of xenografts of human SK-N-(BE)2 cells with amplified MYCN was suppressed by oral administration of opaganib at doses that are well tolerated in mice. Combining opaganib with temozolomide plus irinotecan, considered the backbone for therapy of relapsed or refractory NB, resulted in increased antitumor activity in vivo compared with temozolomide plus irinotecan or opaganib alone. Mice did not lose additional weight when opaganib was combined with temozolomide plus irinotecan, indicating that the combination is well tolerated. Opaganib has additive antitumor activity toward Neuro-2a tumors when combined with the checkpoint inhibitor anti-CTLA-4 antibody; however, the combination of opaganib with anti-PD-1 or anti-PD-L1 antibodies did not provide increased antitumor activity over that seen with opaganib alone. Overall, the data demonstrate that opaganib modulates sphingolipid metabolism and intracellular signaling in NB cells and inhibits NB tumor growth alone and in combination with other anticancer drugs. Amplified MYCN does not confer resistance to opaganib, and, in fact, the drug attenuates the expression of both c-Myc and N-Myc. The safety of opaganib has been established in clinical trials with adults with advanced cancer or severe COVID-19, and so opaganib has excellent potential for treating patients with NB, particularly in combination with temozolomide and irinotecan or anti-CTLA-4 antibody.
Collapse
Affiliation(s)
| | | | | | | | - Charles D. Smith
- Apogee Biotechnology Corporation, 1214 Research Blvd, Suite 2015, Hummelstown, PA 17036, USA
| |
Collapse
|
11
|
Di Matteo S, Bilotta MT, Pelosi A, Haas D, Theinert T, Weber G, Schlegel PG, Berg M, Moretta L, Maggi E, Azzarone B, Vacca P, Tumino N, Caruana I. Transition to a mesenchymal state in neuroblastoma may be characterized by a high expression of GD2 and by the acquisition of immune escape from NK cells. Front Immunol 2024; 15:1382931. [PMID: 38736882 PMCID: PMC11082345 DOI: 10.3389/fimmu.2024.1382931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/09/2024] [Indexed: 05/14/2024] Open
Abstract
Background Neuroblastoma (NB) is characterized by both adrenergic (ADRN) and undifferentiated mesenchymal (MES) subsets. The ganglioside sialic acid-containing glycosphingolipid (GD2) is widely overexpressed on tumors of neuroectodermal origin promoting malignant phenotypes. MES cells are greatly enriched in post-therapy and relapsing tumors and are characterized by decreased expression of GD2. This event may cause failure of GD2-based immunotherapy. NK cells represent a key innate cell subset able to efficiently kill tumors. However, the tumor microenvironment (TME) that includes tumor cells and tumor-associated (TA) cells could inhibit their effector function. Methods We studied eight NB primary cultures that, in comparison with commercial cell lines, more faithfully reflect the tumor cell characteristics. We studied four primary NB-MES cell cultures and two pairs of MES/ADRN (691 and 717) primary cultures, derived from the same patient. In particular, in the six human NB primary cultures, we assessed their phenotype, the expression of GD2, and the enzymes that control its expression, as well as their interactions with NK cells, using flow cytometry, RT-qPCR, and cytotoxicity assays. Results We identified mature (CD105+/CD133-) and undifferentiated (CD133+/CD105-) NB subsets that express high levels of the MES transcripts WWTR1 and SIX4. In addition, undifferentiated MES cells display a strong resistance to NK-mediated killing. On the contrary, mature NB-MES cells display an intermediate resistance to NK-mediated killing and exhibit some immunomodulatory capacities on NK cells but do not inhibit their cytolytic activity. Notably, independent from their undifferentiated or mature phenotype, NB-MES cells express GD2 that can be further upregulated in undifferentiated NB-MES cells upon co-culture with NK cells, leading to the generation of mature mesenchymal GD2bright neuroblasts. Concerning 691 and 717, they show high levels of GD2 and resistance to NK cell-mediated killing that can be overcome by the administration of dinutuximab beta, the anti-GD2 monoclonal antibody applied in the clinic. Conclusions NB is a heterogeneous tumor representing a further hurdle in NB immunotherapy. However, different from what was reported with NB commercial cells and independent of their MES/ADRN phenotype, the expression of GD2 and its displayed sensitivity to anti-GD2 mAb ADCC indicated the possible effectiveness of anti-GD2 immunotherapy.
Collapse
Affiliation(s)
- Sabina Di Matteo
- Tumour Immunology Unit, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Maria Teresa Bilotta
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Pelosi
- Tumour Immunology Unit, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Dorothee Haas
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Tobias Theinert
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Gerrit Weber
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Paul-Gerhardt Schlegel
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Matthias Berg
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| | - Lorenzo Moretta
- Tumour Immunology Unit, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Enrico Maggi
- Tumour Immunology Unit, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Bruno Azzarone
- Tumour Immunology Unit, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Paola Vacca
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Nicola Tumino
- Innate Lymphoid Cells Unit, Immunology Research Area, Bambino Gesù Children’s Hospital Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Ignazio Caruana
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
12
|
Zheng M, Kumar A, Sharma V, Behl T, Sehgal A, Wal P, Shinde NV, Kawaduji BS, Kapoor A, Anwer MK, Gulati M, Shen B, Singla RK, Bungau SG. Revolutionizing pediatric neuroblastoma treatment: unraveling new molecular targets for precision interventions. Front Cell Dev Biol 2024; 12:1353860. [PMID: 38601081 PMCID: PMC11004261 DOI: 10.3389/fcell.2024.1353860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/13/2024] [Indexed: 04/12/2024] Open
Abstract
Neuroblastoma (NB) is the most frequent solid tumor in pediatric cases, contributing to around 15% of childhood cancer-related deaths. The wide-ranging genetic, morphological, and clinical diversity within NB complicates the success of current treatment methods. Acquiring an in-depth understanding of genetic alterations implicated in the development of NB is essential for creating safer and more efficient therapies for this severe condition. Several molecular signatures are being studied as potential targets for developing new treatments for NB patients. In this article, we have examined the molecular factors and genetic irregularities, including those within insulin gene enhancer binding protein 1 (ISL1), dihydropyrimidinase-like 3 (DPYSL3), receptor tyrosine kinase-like orphan receptor 1 (ROR1) and murine double minute 2-tumor protein 53 (MDM2-P53) that play an essential role in the development of NB. A thorough summary of the molecular targeted treatments currently being studied in pre-clinical and clinical trials has been described. Recent studies of immunotherapeutic agents used in NB are also studied in this article. Moreover, we explore potential future directions to discover new targets and treatments to enhance existing therapies and ultimately improve treatment outcomes and survival rates for NB patients.
Collapse
Affiliation(s)
- Min Zheng
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ankush Kumar
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Vishakha Sharma
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali, Punjab, India
| | - Aayush Sehgal
- GHG Khalsa College of Pharmacy, Ludhiana, Punjab, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, Uttar Pradesh, India
| | | | | | - Anupriya Kapoor
- School of Pharmaceutical Sciences, Chhatrapati Shahu Ji Maharaj University, Kanpur, Uttar Pradesh, India
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Australian Research Consortium in Complementary and Integrative Medicine, Faculty of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K. Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
- Doctoral School of Biomedical Sciences, University of Oradea, Oradea, Romania
| |
Collapse
|
13
|
Kwon Y, Choi Y, Kim M, Jo H, Jeong MS, Jung HS, Jeoung D. HDAC6-MYCN-CXCL3 axis mediates allergic inflammation and is necessary for allergic inflammation-promoted cellular interactions. Mol Immunol 2024; 166:1-15. [PMID: 38176167 DOI: 10.1016/j.molimm.2023.12.007] [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: 09/01/2023] [Revised: 11/27/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024]
Abstract
Histone deacetylase 6 (HDAC6) has been shown to play an important role in allergic inflammation. This study hypothesized that novel downstream targets of HDAC6 would mediate allergic inflammation. Experiments employing HDAC6 knock out C57BL/6 mice showed that HDAC6 mediated passive cutaneous anaphylaxis (PCA) and passive systemic anaphylaxis (PSA). Antigen stimulation increased expression of N-myc (MYCN) and CXCL3 in an HDAC6-dependent manner in the bone marrow-derived mast cells. MYCN and CXCL3 were necessary for both PCA and PSA. The role of early growth response 3 (EGR3) in the regulation of HDAC6 expression has been reported. ChIP assays showed EGR3 as a direct regulator of MYCN. miR-34a-5p was predicted to be a negative regulator of MYCN. Luciferase activity assays showed miR-34a-5p as a direct regulator of MYCN. miR-34a-5p mimic negatively regulated PCA and PSA. MYCN decreased miR-34a-5p expression in antigen-stimulated rat basophilic leukemia cells (RBL2H3). MYCN was shown to bind to the promoter sequence of CXCL3. In an IgE-independent manner, recombinant CXCL3 protein increased expression of HDAC6, MYCN, and β-hexosaminidase activity in RBL2H3 cells. Mouse recombinant CXCL3 protein enhanced the angiogenic potential of the culture medium of RBL2H3. CXCL3 was necessary for the enhanced angiogenic potential of the culture medium of antigen-stimulated RBL2H3. The culture medium of RBL2H3 was able to induce M2 macrophage polarization in a CXCL3-dependent manner. Recombinant CXCL3 protein also increased the expression of markers of M2 macrophage. Thus, the identification of the novel role of HDAC6-MYCN-CXCL3 axis can help better understand the pathogenesis of anaphylaxis.
Collapse
Affiliation(s)
- Yoojung Kwon
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Yunji Choi
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Misun Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, South Korea
| | - Hyein Jo
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Myeong Seon Jeong
- Chuncheon Center, Korea Basic Science Institute, Chuncheon 24341, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, South Korea.
| |
Collapse
|
14
|
Stip MC, Teeuwen L, Dierselhuis MP, Leusen JHW, Krijgsman D. Targeting the myeloid microenvironment in neuroblastoma. J Exp Clin Cancer Res 2023; 42:337. [PMID: 38087370 PMCID: PMC10716967 DOI: 10.1186/s13046-023-02913-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Myeloid cells (granulocytes and monocytes/macrophages) play an important role in neuroblastoma. By inducing a complex immunosuppressive network, myeloid cells pose a challenge for the adaptive immune system to eliminate tumor cells, especially in high-risk neuroblastoma. This review first summarizes the pro- and anti-tumorigenic functions of myeloid cells, including granulocytes, monocytes, macrophages, and myeloid-derived suppressor cells (MDSC) during the development and progression of neuroblastoma. Secondly, we discuss how myeloid cells are engaged in the current treatment regimen and explore novel strategies to target these cells in neuroblastoma. These strategies include: (1) engaging myeloid cells as effector cells, (2) ablating myeloid cells or blocking the recruitment of myeloid cells to the tumor microenvironment and (3) reprogramming myeloid cells. Here we describe that despite their immunosuppressive traits, tumor-associated myeloid cells can still be engaged as effector cells, which is clear in anti-GD2 immunotherapy. However, their full potential is not yet reached, and myeloid cell engagement can be enhanced, for example by targeting the CD47/SIRPα axis. Though depletion of myeloid cells or blocking myeloid cell infiltration has been proven effective, this strategy also depletes possible effector cells for immunotherapy from the tumor microenvironment. Therefore, reprogramming of suppressive myeloid cells might be the optimal strategy, which reverses immunosuppressive traits, preserves myeloid cells as effectors of immunotherapy, and subsequently reactivates tumor-infiltrating T cells.
Collapse
Affiliation(s)
- Marjolein C Stip
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Loes Teeuwen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | | | - Jeanette H W Leusen
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Daniëlle Krijgsman
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
- Center for Molecular Medicine, University Medical Center Utrecht, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
15
|
Deng Z, Richardson DR. The Myc Family and the Metastasis Suppressor NDRG1: Targeting Key Molecular Interactions with Innovative Therapeutics. Pharmacol Rev 2023; 75:1007-1035. [PMID: 37280098 DOI: 10.1124/pharmrev.122.000795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/07/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Cancer is a leading cause of death worldwide, resulting in ∼10 million deaths in 2020. Major oncogenic effectors are the Myc proto-oncogene family, which consists of three members including c-Myc, N-Myc, and L-Myc. As a pertinent example of the role of the Myc family in tumorigenesis, amplification of MYCN in childhood neuroblastoma strongly correlates with poor patient prognosis. Complexes between Myc oncoproteins and their partners such as hypoxia-inducible factor-1α and Myc-associated protein X (MAX) result in proliferation arrest and pro-proliferative effects, respectively. Interactions with other proteins are also important for N-Myc activity. For instance, the enhancer of zest homolog 2 (EZH2) binds directly to N-Myc to stabilize it by acting as a competitor against the ubiquitin ligase, SCFFBXW7, which prevents proteasomal degradation. Heat shock protein 90 may also be involved in N-Myc stabilization since it binds to EZH2 and prevents its degradation. N-Myc downstream-regulated gene 1 (NDRG1) is downregulated by N-Myc and participates in the regulation of cellular proliferation via associating with other proteins, such as glycogen synthase kinase-3β and low-density lipoprotein receptor-related protein 6. These molecular interactions provide a better understanding of the biologic roles of N-Myc and NDRG1, which can be potentially used as therapeutic targets. In addition to directly targeting these proteins, disrupting their key interactions may also be a promising strategy for anti-cancer drug development. This review examines the interactions between the Myc proteins and other molecules, with a special focus on the relationship between N-Myc and NDRG1 and possible therapeutic interventions. SIGNIFICANCE STATEMENT: Neuroblastoma is one of the most common childhood solid tumors, with a dismal five-year survival rate. This problem makes it imperative to discover new and more effective therapeutics. The molecular interactions between major oncogenic drivers of the Myc family and other key proteins; for example, the metastasis suppressor, NDRG1, may potentially be used as targets for anti-neuroblastoma drug development. In addition to directly targeting these proteins, disrupting their key molecular interactions may also be promising for drug discovery.
Collapse
Affiliation(s)
- Zhao Deng
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| |
Collapse
|
16
|
Kaufman ME, Vayani OR, Moore K, Chlenski A, Wu T, Chavez G, Lee SM, Desai AV, He C, Cohn SL, Applebaum MA. T-cell inflammation is prognostic of survival in patients with high-risk neuroblastoma enriched for an adrenergic signature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546541. [PMID: 37425883 PMCID: PMC10326980 DOI: 10.1101/2023.06.26.546541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Purpose T-cell inflammation (TCI) has been shown to be a prognostic marker in neuroblastoma, a tumor comprised of cells that can exist in two epigenetic states, adrenergic (ADRN) and mesenchymal (MES). We hypothesized that elucidating unique and overlapping aspects of these biologic features could serve as novel biomarkers. Patients and Methods We detected lineage-specific, single-stranded super-enhancers defining ADRN and MES specific genes. Publicly available neuroblastoma RNA-seq data from GSE49711 (Cohort 1) and TARGET (Cohort 2) were assigned MES, ADRN, and TCI scores. Tumors were characterized as MES (top 33%) or ADRN (bottom 33%), and TCI (top 67% TCI score) or non-inflamed (bottom 33% TCI score). Overall survival (OS) was assessed using the Kaplan-Meier method, and differences were assessed by the log-rank test. Results We identified 159 MES genes and 373 ADRN genes. TCI scores were correlated with MES scores (R=0.56, p<0.001 and R=0.38, p<0.001) and anticorrelated with MYCN -amplification (R=-0.29, p<0.001 and -0.18, p=0.03) in both cohorts. Among Cohort 1 patients with high-risk, ADRN tumors (n=59), those with TCI tumors (n=22) had superior OS to those with non-inflammed tumors (n=37) (p=0.01), though this comparison did not reach significance in Cohort 2. TCI status was not associated with survival in patients with high-risk MES tumors in either cohort. Conclusions High inflammation scores were correlated with improved survival in some high-risk patients with, ADRN but not MES neuroblastoma. These findings have implications for approaches to treating high-risk neuroblastoma.
Collapse
|
17
|
Vitale C, Bottino C, Castriconi R. Monocyte and Macrophage in Neuroblastoma: Blocking Their Pro-Tumoral Functions and Strengthening Their Crosstalk with Natural Killer Cells. Cells 2023; 12:885. [PMID: 36980226 PMCID: PMC10047506 DOI: 10.3390/cells12060885] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Over the past decade, immunotherapy has represented an enormous step forward in the fight against cancer. Immunotherapeutic approaches have increasingly become a fundamental part of the combined therapies currently adopted in the treatment of patients with high-risk (HR) neuroblastoma (NB). An increasing number of studies focus on the understanding of the immune landscape in NB and, since this tumor expresses low or null levels of MHC class I, on the development of new strategies aimed at enhancing innate immunity, especially Natural Killer (NK) cells and macrophages. There is growing evidence that, within the NB tumor microenvironment (TME), tumor-associated macrophages (TAMs), which mainly present an M2-like phenotype, have a crucial role in mediating NB development and immune evasion, and they have been correlated to poor clinical outcomes. Importantly, TAM can also impair the antibody-dependent cellular cytotoxicity (ADCC) mediated by NK cells upon the administration of anti-GD2 monoclonal antibodies (mAbs), the current standard immunotherapy for HR-NB patients. This review deals with the main mechanisms regulating the crosstalk among NB cells and TAMs or other cellular components of the TME, which support tumor development and induce drug resistance. Furthermore, we will address the most recent strategies aimed at limiting the number of pro-tumoral macrophages within the TME, reprogramming the TAMs functional state, thus enhancing NK cell functions. We also prospectively discuss new or unexplored aspects of human macrophage heterogeneity.
Collapse
Affiliation(s)
- Chiara Vitale
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
- Laboratory of Clinical and Experimental Immunology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| |
Collapse
|
18
|
Antigene MYCN Silencing by BGA002 Inhibits SCLC Progression Blocking mTOR Pathway and Overcomes Multidrug Resistance. Cancers (Basel) 2023; 15:cancers15030990. [PMID: 36765949 PMCID: PMC9913109 DOI: 10.3390/cancers15030990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Small-cell lung cancer (SCLC) is the most aggressive lung cancer type, and is associated with smoking, low survival rate due to high vascularization, metastasis and drug resistance. Alterations in MYC family members are biomarkers of poor prognosis for a large number of SCLC. In particular, MYCN alterations define SCLC cases with immunotherapy failure. MYCN has a highly restricted pattern of expression in normal cells and is an ideal target for cancer therapy but is undruggable by traditional approaches. We propose an innovative approach to MYCN inhibition by an MYCN-specific antigene-PNA oligonucleotide (BGA002)-as a new precision medicine for MYCN-related SCLC. We found that BGA002 profoundly and specifically inhibited MYCN expression in SCLC cells, leading to cell-growth inhibition and apoptosis, while also overcoming multidrug resistance. These effects are driven by mTOR pathway block in concomitance with autophagy reactivation, thus avoiding the side effects of targeting mTOR in healthy cells. Moreover, we identified an MYCN-related SCLC gene signature comprehending CNTFR, DLX5 and TNFAIP3, that was reverted by BGA002. Finally, systemic treatment with BGA002 significantly increased survival in MYCN-amplified SCLC mouse models, including in a multidrug-resistant model in which tumor vascularization was also eliminated. These findings warrant the clinical testing of BGA002 in MYCN-related SCLC.
Collapse
|
19
|
Halliwell E, Vitali A, Muller H, Alonso-Ferrero M, Barisa M, Gavriil A, Piapi A, Leboreiro-Babe C, Gileadi T, Yeung J, Pataillot-Meakin T, Fisher J, Tucker L, Donovan L, Chesler L, Chester K, Anderson J. Targeting of low ALK antigen density neuroblastoma using AND logic-gate engineered CAR-T cells. Cytotherapy 2023; 25:46-58. [PMID: 36396552 DOI: 10.1016/j.jcyt.2022.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND AIMS The targeting of solid cancers with chimeric antigen receptor (CAR) T cells faces many technological hurdles, including selection of optimal target antigens. Promising pre-clinical and clinical data of CAR T-cell activity have emerged from targeting surface antigens such as GD2 and B7H3 in childhood cancer neuroblastoma. Anaplastic lymphoma kinase (ALK) is expressed in a majority of neuroblastomas at low antigen density but is largely absent from healthy tissues. METHODS To explore an alternate target antigen for neuroblastoma CAR T-cell therapy, the authors generated and screened a single-chain variable fragment library targeting ALK extracellular domain to make a panel of new anti-ALK CAR T-cell constructs. RESULTS A lead novel CAR T-cell construct was capable of specific cytotoxicity against neuroblastoma cells expressing low levels of ALK, but with only weak cytokine and proliferative T-cell responses. To explore strategies for amplifying ALK CAR T cells, the authors generated a co-CAR approach in which T cells received signal 1 from a first-generation ALK construct and signal 2 from anti-B7H3 or GD2 chimeric co-stimulatory receptors. The co-CAR approach successfully demonstrated the ability to avoid targeting single-antigen-positive targets as a strategy for mitigating on-target off-tumor toxicity. CONCLUSIONS These data provide further proof of concept for ALK as a neuroblastoma CAR T-cell target.
Collapse
Affiliation(s)
- Emma Halliwell
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alice Vitali
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Henrike Muller
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Marta Barisa
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Artemis Gavriil
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alice Piapi
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Talia Gileadi
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Jenny Yeung
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK; UCL Cancer Institute, London, UK
| | | | - Jonathan Fisher
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Laura Donovan
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - John Anderson
- Cancer Section, UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
20
|
Valind A, Verhoeven BM, Enoksson J, Karlsson J, Christensson G, Mañas A, Aaltonen K, Jansson C, Bexell D, Baryawno N, Gisselsson D, Hagerling C. Macrophage infiltration promotes regrowth in MYCN-amplified neuroblastoma after chemotherapy. Oncoimmunology 2023; 12:2184130. [PMID: 36875552 PMCID: PMC9980604 DOI: 10.1080/2162402x.2023.2184130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Despite aggressive treatment, the 5-year event-free survival rate for children with high-risk neuroblastoma is <50%. While most high-risk neuroblastoma patients initially respond to treatment, often with complete clinical remission, many eventually relapse with therapy-resistant tumors. Novel therapeutic alternatives that prevent the recurrence of therapy-resistant tumors are urgently needed. To understand the adaptation of neuroblastoma under therapy, we analyzed the transcriptomic landscape in 46 clinical tumor samples collected before (PRE) or after (POST) treatment from 22 neuroblastoma patients. RNA sequencing revealed that many of the top-upregulated biological processes in POST MYCN amplified (MNA+) tumors compared to PRE MNA+ tumors were immune-related, and there was a significant increase in numerous genes associated with macrophages. The infiltration of macrophages was corroborated by immunohistochemistry and spatial digital protein profiling. Moreover, POST MNA+ tumor cells were more immunogenic compared to PRE MNA+ tumor cells. To find support for the macrophage-induced outgrowth of certain subpopulations of immunogenic tumor cells following treatment, we examined the genetic landscape in multiple clinical PRE and POST tumor samples from nine neuroblastoma patients revealing a significant correlation between an increased amount of copy number aberrations (CNA) and macrophage infiltration in POST MNA+ tumor samples. Using an in vivo neuroblastoma patient-derived xenograft (PDX) chemotherapy model, we further show that inhibition of macrophage recruitment with anti-CSF1R treatment prevents the regrowth of MNA+ tumors following chemotherapy. Taken together, our work supports a therapeutic strategy for fighting the relapse of MNA+ neuroblastoma by targeting the immune microenvironment.
Collapse
Affiliation(s)
- Anders Valind
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Sweden Karolinska Institute, Lund, Sweden.,Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Bronte Manouk Verhoeven
- Childhood Cancer Research Unit, Department of Women's and Children's Healthy, Karolinska Institute, Stockholm, Sweden
| | - Jens Enoksson
- Department of Pathology, Laboratory Medicine, Skåne University Hospital, Lund, Sweden
| | - Jenny Karlsson
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Sweden Karolinska Institute, Lund, Sweden
| | - Gustav Christensson
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Sweden Karolinska Institute, Lund, Sweden
| | - Adriana Mañas
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kristina Aaltonen
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Caroline Jansson
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Sweden Karolinska Institute, Lund, Sweden
| | - Daniel Bexell
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women's and Children's Healthy, Karolinska Institute, Stockholm, Sweden
| | - David Gisselsson
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Sweden Karolinska Institute, Lund, Sweden.,Department of Pathology, Laboratory Medicine, Skåne University Hospital, Lund, Sweden
| | - Catharina Hagerling
- Department of Laboratory Medicine, Division of Clinical Genetics, Lund University, Sweden Karolinska Institute, Lund, Sweden.,Department of Pathology, Laboratory Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
21
|
Qiao J, Liu J, Jacobson JC, Clark RA, Lee S, Liu L, An Z, Zhang N, Chung DH. Anti-GRP-R monoclonal antibody antitumor therapy against neuroblastoma. PLoS One 2022; 17:e0277956. [PMID: 36525420 PMCID: PMC9757561 DOI: 10.1371/journal.pone.0277956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/07/2022] [Indexed: 12/23/2022] Open
Abstract
Standard treatment for patients with high-risk neuroblastoma remains multimodal therapy including chemoradiation, surgical resection, and autologous stem cell rescue. Immunotherapy has demonstrated success in treating many types of cancers; however, its use in pediatric solid tumors has been limited by low tumor mutation burdens. Gastrin-releasing peptide receptor (GRP-R) is overexpressed in numerous malignancies, including poorly-differentiated neuroblastoma. Monoclonal antibodies (mAbs) to GRP-R have yet to be developed but could serve as a potential novel immunotherapy. This preclinical study aims to evaluate the efficacy of a novel GRP-R mAb immunotherapy against neuroblastoma. We established four candidate anti-GRP-R mAbs by screening a single-chain variable fragment (scFv) library. GRP-R mAb-1 demonstrated the highest efficacy with the lowest EC50 at 4.607 ng/ml against GRP-R expressing neuroblastoma cells, blocked the GRP-ligand activation of GRP-R and its downstream PI3K/AKT signaling. This resulted in functional inhibition of cell proliferation and anchorage-independent growth, indicating that mAb-1 has an antagonist inhibitory role on GRP-R. To examine the antibody-dependent cellular cytotoxicity (ADCC) of GRP-R mAb-1 on neuroblastoma, we co-cultured neuroblastoma cells with natural killer (NK) cells versus GRP-R mAb-1 treatment alone. GRP-R mAb-1 mediated ADCC effects on neuroblastoma cells and induced release of IFNγ by NK cells under co-culture conditions in vitro. The cytotoxic effects of mAb-1 were confirmed with the secretion of cytotoxic granzyme B from NK cells and the reduction of mitotic tumor cells in vivo using a murine tumor xenograft model. In summary, GRP-R mAb-1 demonstrated efficacious anti-tumor effects on neuroblastoma cells in preclinical models. Importantly, GRP-R mAb-1 may be an efficacious, novel immunotherapy in the treatment of high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Jingbo Qiao
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Junquan Liu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Jillian C. Jacobson
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Rachael A. Clark
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Sora Lee
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Li Liu
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dai H. Chung
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Surgery, Children’s Health, Dallas, Texas, United States of America
| |
Collapse
|
22
|
Ho GY, Kyran EL, Bedo J, Wakefield MJ, Ennis DP, Mirza HB, Vandenberg CJ, Lieschke E, Farrell A, Hadla A, Lim R, Dall G, Vince JE, Chua NK, Kondrashova O, Upstill-Goddard R, Bailey UM, Dowson S, Roxburgh P, Glasspool RM, Bryson G, Biankin AV, Cooke SL, Ratnayake G, McNally O, Traficante N, DeFazio A, Weroha SJ, Bowtell DD, McNeish IA, Papenfuss AT, Scott CL, Barker HE. Epithelial-to-Mesenchymal Transition Supports Ovarian Carcinosarcoma Tumorigenesis and Confers Sensitivity to Microtubule Targeting with Eribulin. Cancer Res 2022; 82:4457-4473. [PMID: 36206301 PMCID: PMC9716257 DOI: 10.1158/0008-5472.can-21-4012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 06/15/2022] [Accepted: 10/04/2022] [Indexed: 01/24/2023]
Abstract
Ovarian carcinosarcoma (OCS) is an aggressive and rare tumor type with limited treatment options. OCS is hypothesized to develop via the combination theory, with a single progenitor resulting in carcinomatous and sarcomatous components, or alternatively via the conversion theory, with the sarcomatous component developing from the carcinomatous component through epithelial-to-mesenchymal transition (EMT). In this study, we analyzed DNA variants from isolated carcinoma and sarcoma components to show that OCS from 18 women is monoclonal. RNA sequencing indicated that the carcinoma components were more mesenchymal when compared with pure epithelial ovarian carcinomas, supporting the conversion theory and suggesting that EMT is important in the formation of these tumors. Preclinical OCS models were used to test the efficacy of microtubule-targeting drugs, including eribulin, which has previously been shown to reverse EMT characteristics in breast cancers and induce differentiation in sarcomas. Vinorelbine and eribulin more effectively inhibited OCS growth than standard-of-care platinum-based chemotherapy, and treatment with eribulin reduced mesenchymal characteristics and N-MYC expression in OCS patient-derived xenografts. Eribulin treatment resulted in an accumulation of intracellular cholesterol in OCS cells, which triggered a downregulation of the mevalonate pathway and prevented further cholesterol biosynthesis. Finally, eribulin increased expression of genes related to immune activation and increased the intratumoral accumulation of CD8+ T cells, supporting exploration of immunotherapy combinations in the clinic. Together, these data indicate that EMT plays a key role in OCS tumorigenesis and support the conversion theory for OCS histogenesis. Targeting EMT using eribulin could help improve OCS patient outcomes. SIGNIFICANCE Genomic analyses and preclinical models of ovarian carcinosarcoma support the conversion theory for disease development and indicate that microtubule inhibitors could be used to suppress EMT and stimulate antitumor immunity.
Collapse
Affiliation(s)
- Gwo Yaw Ho
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Elizabeth L. Kyran
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Justin Bedo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- School of Computing and Information Systems, the University of Melbourne, Parkville, Victoria, Australia
| | - Matthew J. Wakefield
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
| | - Darren P. Ennis
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Hasan B. Mirza
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Cassandra J. Vandenberg
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth Lieschke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew Farrell
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Anthony Hadla
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ratana Lim
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Genevieve Dall
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - James E. Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ngee Kiat Chua
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Olga Kondrashova
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Rosanna Upstill-Goddard
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Ulla-Maja Bailey
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Suzanne Dowson
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Patricia Roxburgh
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Rosalind M. Glasspool
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Gareth Bryson
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Andrew V. Biankin
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | | | - Susanna L. Cooke
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | | | - Orla McNally
- The Royal Women's Hospital, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Nadia Traficante
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, Australia
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, Australia
| | - S. John Weroha
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - David D. Bowtell
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Iain A. McNeish
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Anthony T. Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- The Royal Women's Hospital, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Holly E. Barker
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
23
|
Bartolucci D, Montemurro L, Raieli S, Lampis S, Pession A, Hrelia P, Tonelli R. MYCN Impact on High-Risk Neuroblastoma: From Diagnosis and Prognosis to Targeted Treatment. Cancers (Basel) 2022; 14:4421. [PMID: 36139583 PMCID: PMC9496712 DOI: 10.3390/cancers14184421] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Among childhood cancers, neuroblastoma is the most diffuse solid tumor and the deadliest in children. While to date, the pathology has become progressively manageable with a significant increase in 5-year survival for its less aggressive form, high-risk neuroblastoma (HR-NB) remains a major issue with poor outcome and little survivability of patients. The staging system has also been improved to better fit patient needs and to administer therapies in a more focused manner in consideration of pathology features. New and improved therapies have been developed; nevertheless, low efficacy and high toxicity remain a staple feature of current high-risk neuroblastoma treatment. For this reason, more specific procedures are required, and new therapeutic targets are also needed for a precise medicine approach. In this scenario, MYCN is certainly one of the most interesting targets. Indeed, MYCN is one of the most relevant hallmarks of HR-NB, and many studies has been carried out in recent years to discover potent and specific inhibitors to block its activities and any related oncogenic function. N-Myc protein has been considered an undruggable target for a long time. Thus, many new indirect and direct approaches have been discovered and preclinically evaluated for the interaction with MYCN and its pathways; a few of the most promising approaches are nearing clinical application for the investigation in HR-NB.
Collapse
Affiliation(s)
| | - Luca Montemurro
- Pediatric Oncology and Hematology Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | | | | | - Andrea Pession
- Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Roberto Tonelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
24
|
Bartolucci D, Pession A, Hrelia P, Tonelli R. Precision Anti-Cancer Medicines by Oligonucleotide Therapeutics in Clinical Research Targeting Undruggable Proteins and Non-Coding RNAs. Pharmaceutics 2022; 14:pharmaceutics14071453. [PMID: 35890348 PMCID: PMC9315662 DOI: 10.3390/pharmaceutics14071453] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/24/2022] [Accepted: 07/08/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer incidence and mortality continue to increase, while the conventional chemotherapeutic drugs confer limited efficacy and relevant toxic side effects. Novel strategies are urgently needed for more effective and safe therapeutics in oncology. However, a large number of proteins are considered undruggable by conventional drugs, such as the small molecules. Moreover, the mRNA itself retains oncological functions, and its targeting offers the double advantage of blocking the tumorigenic activities of the mRNA and the translation into protein. Finally, a large family of non-coding RNAs (ncRNAs) has recently emerged that are also dysregulated in cancer, but they could not be targeted by drugs directed against the proteins. In this context, this review describes how the oligonucleotide therapeutics targeting RNA or DNA sequences, are emerging as a new class of drugs, able to tackle the limitations described above. Numerous clinical trials are evaluating oligonucleotides for tumor treatment, and in the next few years some of them are expected to reach the market. We describe the oligonucleotide therapeutics targeting undruggable proteins (focusing on the most relevant, such as those originating from the MYC and RAS gene families), and for ncRNAs, in particular on those that are under clinical trial evaluation in oncology. We highlight the challenges and solutions for the clinical success of oligonucleotide therapeutics, with particular emphasis on the peculiar challenges that render it arduous to treat tumors, such as heterogeneity and the high mutation rate. In the review are presented these and other advantages offered by the oligonucleotide as an emerging class of biotherapeutics for a new era of precision anti-cancer medicine.
Collapse
Affiliation(s)
| | - Andrea Pession
- Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Roberto Tonelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
- Correspondence:
| |
Collapse
|
25
|
Kacher J, Manches O, Aspord C, Sartelet H, Chaperot L. Impaired Antitumor Immune Response in MYCN-amplified Neuroblastoma Is Associated with Lack of CCL2 Secretion and Poor Dendritic Cell Recruitment. CANCER RESEARCH COMMUNICATIONS 2022; 2:577-589. [PMID: 36923280 PMCID: PMC10010397 DOI: 10.1158/2767-9764.crc-21-0134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/28/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022]
Abstract
In neuroblastoma, MYCN amplification is associated with sparse immune infiltrate and poor prognosis. Dendritic cells (DC) are crucial immune sentinels but their involvement in neuroblastoma pathogenesis is poorly understood. We observed that the migration of monocytes, myeloid and plasmacytoid DC induced by MYCN-nonamplified neuroblastoma supernatants was abrogated by the addition of anti-CCL2 antibodies, demonstrating the involvement of the CCR2/CCL2 axis in their recruitment by these tumors. Using public RNA sequencing and microarray datasets, we describe lower level of expression of CCL2 in MYCN-amplified neuroblastoma tumors, and we propose a working model for T-cell recruitment in neuroblastoma tumors in which CCL2 produced by neuroblastoma cells initiates the recruitment of monocytes, myeloid and plasmacytoid DCs. Among these cells, the CD1c+ subset may recruit T cells by means of CCL19/CCL22 secretion. In vitro, supernatants from DCs cocultured with neuroblastoma cell lines and activated contain CCL22 and CCL19, and are chemotactic for both CD4+ and CD8+ T cells. We also looked at immunomodulation induced by neuroblastoma cell lines, and found MYCN-nonamplified neuroblastoma cell lines were able to create a microenvironment where DC activation is enhanced. Overall, our findings highlight a major role for CCL2/CCR2 axis in monocytes, myeloid and plasmacytoid cells recruitment toward MYCN-nonamplified neuroblastoma, allowing further immune cell recruitment, and show that these tumors present a microenvironment that can favor DC responses. Significance In MYCN-nonamplified neuroblastoma, CCL2 produced by neuroblastoma cells induces the recruitment of antigen-presenting cells (DCs and monocytes/macrophages), allowing infiltration by T cells, in link with CCL19 and CCL22 production, hence favoring immune responses.
Collapse
Affiliation(s)
- Jamila Kacher
- Institute for Advanced Biosciences, Inserm U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France.,Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Olivier Manches
- Institute for Advanced Biosciences, Inserm U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France.,Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Caroline Aspord
- Institute for Advanced Biosciences, Inserm U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France.,Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Hervé Sartelet
- Laboratoire de Biopathologie, CHRU de Nancy, Nancy, France.,Inserm U1256, Université de Lorraine, Nancy, France
| | - Laurence Chaperot
- Institute for Advanced Biosciences, Inserm U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France.,Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| |
Collapse
|
26
|
Lampis S, Raieli S, Montemurro L, Bartolucci D, Amadesi C, Bortolotti S, Angelucci S, Scardovi AL, Nieddu G, Cerisoli L, Paganelli F, Valente S, Fischer M, Martelli AM, Pasquinelli G, Pession A, Hrelia P, Tonelli R. The MYCN inhibitor BGA002 restores the retinoic acid response leading to differentiation or apoptosis by the mTOR block in MYCN-amplified neuroblastoma. J Exp Clin Cancer Res 2022; 41:160. [PMID: 35490242 PMCID: PMC9055702 DOI: 10.1186/s13046-022-02367-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/18/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Neuroblastoma is a deadly childhood cancer, and MYCN-amplified neuroblastoma (MNA-NB) patients have the worst prognoses and are therapy-resistant. While retinoic acid (RA) is beneficial for some neuroblastoma patients, the cause of RA resistance is unknown. Thus, there remains a need for new therapies to treat neuroblastoma. Here we explored the possibility of combining a MYCN-specific antigene oligonucleotide BGA002 and RA as therapeutic approach to restore sensitivity to RA in NB. METHODS By molecular and cellular biology techniques, we assessed the combined effect of the two compounds in NB cell lines and in a xenograft mouse model MNA-NB. RESULTS We found that MYCN-specific inhibition by BGA002 in combination with RA (BGA002-RA) act synergistically and overcame resistance in NB cell lines. BGA002-RA also reactivated neuron differentiation (or led to apoptosis) and inhibited invasiveness capacity in MNA-NB. Moreover, we found that neuroblastoma had the highest level of mRNA expression of mTOR pathway genes, and that BGA002 led to mTOR pathway inhibition followed by autophagy reactivation in MNA-NB cells, which was strengthened by BGA002-RA. BGA002-RA in vivo treatment also eliminated tumor vascularization in a MNA-NB mouse model and significantly increased survival. CONCLUSION Taken together, MYCN modulation mediates the therapeutic efficacy of RA and the development of RA resistance in MNA-NB. Furthermore, by targeting MYCN, a cancer-specific mTOR pathway inhibition occurs only in MNA-NB, thus avoiding the side effects of targeting mTOR in normal cells. These findings warrant clinical testing of BGA002-RA as a strategy for overcoming RA resistance in MNA-NB.
Collapse
Affiliation(s)
| | | | - Luca Montemurro
- Pediatric Unit, S. Orsola IRCCS, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | - Francesca Paganelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- CNR Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza", Unit of Bologna, Bologna, Italy
| | - Sabrina Valente
- Biotechnology and Methods in Laboratory Medicine, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Medical Faculty, Cologne, Germany; and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | | | - Gianandrea Pasquinelli
- Biotechnology and Methods in Laboratory Medicine, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Subcellular nephro-vascular diagnostic program, Pathology Unit S. Orsola IRCCS, University of Bologna, Bologna, Italy
| | - Andrea Pession
- Pediatric Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Roberto Tonelli
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| |
Collapse
|
27
|
Shrestha S, Morcavallo A, Gorrini C, Chesler L. Biological Role of MYCN in Medulloblastoma: Novel Therapeutic Opportunities and Challenges Ahead. Front Oncol 2021; 11:694320. [PMID: 34195095 PMCID: PMC8236857 DOI: 10.3389/fonc.2021.694320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
The constitutive and dysregulated expression of the transcription factor MYCN has a central role in the pathogenesis of the paediatric brain tumour medulloblastoma, with an increased expression of this oncogene correlating with a worse prognosis. Consequently, the genomic and functional alterations of MYCN represent a major therapeutic target to attenuate tumour growth in medulloblastoma. This review will provide a comprehensive synopsis of the biological role of MYCN and its family components, their interaction with distinct signalling pathways, and the implications of this network in medulloblastoma development. We will then summarise the current toolbox for targeting MYCN and highlight novel therapeutic avenues that have the potential to results in better-tailored clinical treatments.
Collapse
Affiliation(s)
- Sumana Shrestha
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom
| | - Alaide Morcavallo
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom
| | - Chiara Gorrini
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom
| | - Louis Chesler
- Division of Clinical Studies, Institute of Cancer Research (ICR), London and Royal Marsden NHS Trust, Sutton, United Kingdom.,Division of Cancer Therapeutics, The Institute of Cancer Research (ICR), and The Royal Marsden NHS Trust, Sutton, United Kingdom
| |
Collapse
|
28
|
Seier JA, Reinhardt J, Saraf K, Ng SS, Layer JP, Corvino D, Althoff K, Giordano FA, Schramm A, Fischer M, Hölzel M. Druggable epigenetic suppression of interferon-induced chemokine expression linked to MYCN amplification in neuroblastoma. J Immunother Cancer 2021; 9:e001335. [PMID: 34016720 PMCID: PMC8141444 DOI: 10.1136/jitc-2020-001335] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Amplification of the MYCN oncogene is a molecular hallmark of aggressive neuroblastoma (NB), a childhood cancer of the sympathetic nervous system. There is evidence that MYCN promotes a non-inflamed and T-cell infiltration-poor ('cold') tumor microenvironment (TME) by suppressing interferon signaling. This may explain, at least in part, why patients with NB seem to have little benefit from single-agent immune checkpoint blockade (ICB) therapy. Targeting MYCN or its effectors could be a strategy to convert a cold TME into a 'hot' (inflamed) TME and improve the efficacy of ICB therapy. METHODS NB transcriptome analyses were used to identify epigenetic drivers of a T-cell infiltration-poor TME. Biological and molecular responses of NB cells to epigenetic drugs and interferon (IFN)-γ exposure were assessed by proliferation assays, immunoblotting, ELISA, qRT-PCR, RNA-seq and ChIP-qPCR as well as co-culture assays with T cells. RESULTS We identified H3K9 euchromatic histone-lysine methyltransferases EHMT2 and EHMT1, also known as G9a and GLP, as epigenetic effectors of the MYCN-driven malignant phenotype and repressors of IFN-γ transcriptional responses in NB cells. EHMT inhibitors enhanced IFN-γ-induced expression of the Th1-type chemokines CXCL9 and CXCL10, key factors of T-cell recruitment into the TME. In MYCN-amplified NB cells, co-inhibition of EZH2 (enhancer of zeste homologue 2), a H3K27 histone methyltransferase cooperating with EHMTs, was needed for strong transcriptional responses to IFN-γ, in line with histone mark changes at CXCL9 and CXCL10 chemokine gene loci. EHMT and EZH2 inhibitor response gene signatures from NB cells were established as surrogate measures and revealed high EHMT and EZH2 activity in MYCN-amplified high-risk NBs with a cold immune phenotype. CONCLUSION Our results delineate a strategy for targeted epigenetic immunomodulation of high-risk NBs, whereby EHMT inhibitors alone or in combination with EZH2 inhibitors (in particular, MYCN-amplified NBs) could promote a T-cell-infiltrated TME via enhanced Th1-type chemokine expression.
Collapse
Affiliation(s)
- Johanna A Seier
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Julia Reinhardt
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Kritika Saraf
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Susanna S Ng
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Julian P Layer
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Department of Radiation Oncology, University Hospital Bonn, Bonn, Germany
| | - Dillon Corvino
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Kristina Althoff
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Frank A Giordano
- Department of Radiation Oncology, University Hospital Bonn, Bonn, Germany
| | - Alexander Schramm
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University Children's Hospital of Cologne, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| |
Collapse
|