1
|
Jiang H, Ye J. The Warburg effect: The hacked mitochondrial-nuclear communication in cancer. Semin Cancer Biol 2025; 112:93-111. [PMID: 40147702 DOI: 10.1016/j.semcancer.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Mitochondrial-nuclear communication is vital for maintaining cellular homeostasis. This communication begins with mitochondria sensing environmental cues and transmitting signals to the nucleus through the retrograde cascade, involving metabolic signals such as substrates for epigenetic modifications, ATP and AMP levels, calcium flux, etc. These signals inform the nucleus about the cell's metabolic state, remodel epigenome and regulate gene expression, and modulate mitochondrial function and dynamics through the anterograde feedback cascade to control cell fate and physiology. Disruption of this communication can lead to cellular dysfunction and disease progression, particularly in cancer. The Warburg effect is the metabolic hallmark of cancer, characterized by disruption of mitochondrial respiration and increased lactate generation from glycolysis. This metabolic reprogramming rewires retrograde signaling, leading to epigenetic changes and dedifferentiation, further reprogramming mitochondrial function and promoting carcinogenesis. Understanding these processes and their link to tumorigenesis is crucial for uncovering tumorigenesis mechanisms. Therapeutic strategies targeting these disrupted pathways, including metabolic and epigenetic components, provide promising avenues for cancer treatment.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Gera S, Guo M, Xie Y, Pollock N, Song M, Weber DR, Denburg M, Zemel B, Mostoufi-Moab S. Assessing bone mineral density and cortical geometry using high-resolution peripheral quantitative computed tomography in pediatric survivors of high-risk neuroblastoma with severe growth failure. Bone 2025; 195:117468. [PMID: 40139336 DOI: 10.1016/j.bone.2025.117468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/21/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
INTRODUCTION Survival of children with high-risk neuroblastoma (HR-NBL) has increased with multimodal therapy. cis-Retinoic acid (cis-RA), cornerstone of HR-NBL therapy, can cause osteoporosis and premature physeal closure. This study utilized high-resolution peripheral quantitative computed tomography (HR-pQCT), for 3D measures of volumetric bone mineral density (BMD) and microarchitecture, to assess impact of HR-NBL therapy on skeletal structure. METHODS We prospectively enrolled 20 HR-NBL survivors and 20 age-, sex-, and race-matched healthy reference participants. We assessed leg lean mass adjusted for leg length by DXA and strength using a Biodex dynamometer. Tibia bone microarchitecture was assessed via HR-pQCT scans at 4 % of tibia length and a cortical site at 30 %. We compared tibia length (cm), cortical and trabecular vBMD (mg HA/cm3), geometric and structural parameters between groups. Linear regression models assessed group differences in bone microarchitecture adjusted for leg lean mass. RESULTS Compared to reference participants, tibia length was significantly shorter in HR-NBL survivors (31.6 cm [27.7,39.5] vs. 36.1 cm [30.4,40], p < 0.005), consistent with significantly lower height Z-score in the HR-NBL cohort (p < 0.001). HR-NBL survivors demonstrated lower cortical area (178.3mm2 [121.9273.5] vs. 214.6mm2 [159.4326.9], p < 0.05) and cortical perimeter (60.0 mm [51.9,82.5] vs. 68.8 mm [57.7,90.8], p < 0.01). After adjusting for tibia length, these differences were no longer significant. Total, cortical, and trabecular volumetric BMD, were not significantly different between groups. Cortical geometry and peak torque deficits were associated with muscle deficits when adjusted for leg lean mass (p < 0.001). CONCLUSION Bone density was not severely impacted in HR-NBL survivors. Muscle deficits persisted years after treatment and underscored cortical geometry deficits.
Collapse
Affiliation(s)
- Sonia Gera
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA
| | - Michelle Guo
- Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Yang Xie
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA
| | - Netanya Pollock
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA
| | - Minkeun Song
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA
| | - David R Weber
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michelle Denburg
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Babette Zemel
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sogol Mostoufi-Moab
- Children's Hospital of Philadelphia, 3500 Civic Center Boulevard, 12th Floor, Philadelphia, PA 19104, USA; Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Simon T, Thole T, Castelli S, Timmermann B, Jazmati D, Schwarz R, Fuchs J, Warmann S, Hubertus J, Schmidt M, Rogasch J, Körber F, Vokuhl C, Schäfer J, Schulte JH, Deubzer H, Rosswog C, Fischer M, Lang P, Langer T, Astrahantseff K, Lode H, Hero B, Eggert A. GPOH Guidelines for Diagnosis and First-line Treatment of Patients with Neuroblastic Tumors, update 2025. KLINISCHE PADIATRIE 2025; 237:117-140. [PMID: 40345224 DOI: 10.1055/a-2556-4302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
The clinical course of neuroblastoma is more heterogeneous than any other malignant disease. Many low-risk patients experience regression after limited or even no chemotherapy. However, more than half of high-risk patients die from disease despite intensive multimodal treatment. Precise disease characterization for each patient at diagnosis is key for risk-adapted treatment. The guidelines presented here incorporate results from national and international clinical trials to produce recommendations for diagnosing and treating neuroblastoma patients in German hospitals outside of clinical trials.
Collapse
Affiliation(s)
- Thorsten Simon
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Theresa Thole
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Sveva Castelli
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Beate Timmermann
- Westgerman Protontherapycenter Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Danny Jazmati
- Department of Radiation Oncology, Heinrich Heine University Düsseldorf, Dusseldorf, Germany
| | | | - Jörg Fuchs
- Pediatric Surgery and Urology, University of Tübingen, Tübingen, Germany
| | - Steven Warmann
- Department of Pediatric Surgery, Charité University Hospital Berlin, Berlin, Germany
| | - Jochen Hubertus
- Department of Pediatric Surgery, Marien-Hospital Witten, Witten, Germany
| | | | - Julian Rogasch
- Nuclear Medicine, Charité University Hospital Berlin, Berlin, Germany
| | - Friederike Körber
- Institut und Poliklinik für Radiologische Diagnostik, Kinderradiologie, University of Cologne, Cologne, Germany
| | - Christian Vokuhl
- Pediatric Pathology, Institute for Pathology, University of Bonn, Bonn, Germany
| | - Jürgen Schäfer
- Diagnostic and Interventional Radiology, University of Tübingen, Tübingen, Germany
| | | | - Hedwig Deubzer
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Carolina Rosswog
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
- Experimental Pediatric Oncology, University of Cologne, Cologne, Germany
| | - Matthias Fischer
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
- Experimental Pediatric Oncology, University of Cologne, Cologne, Germany
| | - Peter Lang
- Pediatric Oncology and Hematology, University of Tübingen, Tübingen, Germany
| | - Thorsten Langer
- Childrens' Hospital, University Hospital Schleswig-Holstein Lübeck Campus, Lübeck, Germany
| | - Kathy Astrahantseff
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| | - Holger Lode
- Pediatric Oncology and Hematology, University of Greifswald, Greifswald, Germany
| | - Barbara Hero
- Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Angelika Eggert
- Pediatric Oncology and Hematology, Charité University Hospital Berlin, Berlin, Germany
| |
Collapse
|
4
|
AlKhazal A, Chohan S, Ross DJ, Kim J, Brown EG. Emerging clinical and research approaches in targeted therapies for high-risk neuroblastoma. Front Oncol 2025; 15:1553511. [PMID: 40104501 PMCID: PMC11913827 DOI: 10.3389/fonc.2025.1553511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/06/2025] [Indexed: 03/20/2025] Open
Abstract
Neuroblastoma is a pediatric cancer that originates from neural crest cells and is the most common extracranial solid tumor in children under five years of age. While low-risk neuroblastoma often regresses spontaneously, high-risk neuroblastoma poses a significant clinical challenge. Recent advances in understanding neuroblastoma's molecular mechanisms have led to the development of targeted therapies that aim to selectively inhibit specific pathways involved in tumor growth and progression, improving patient outcomes while minimizing side effects. This review provides a comprehensive review of neuroblastoma biology and emerging therapeutic strategies. Key topics include (a) immunotherapies and immunotargets, (b) non-coding RNAs (long non-coding RNA, microRNA, and circular RNA), (c) molecular biomarkers and pathways, and (d) limitations and future directions.
Collapse
Affiliation(s)
- Albatool AlKhazal
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Samiha Chohan
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
- Department of Biological Sciences, California State University, Sacramento, Sacramento, CA, United States
| | - Destani J Ross
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Jinhwan Kim
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Erin G Brown
- Department of Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
5
|
Pan M, Zhang Y, Wright WC, Liu X, Passaia B, Currier D, Low J, Chapple RH, Steele JA, Connelly JP, Ju B, Plyler E, Lu M, Loughran AJ, Yang L, Abraham BJ, Pruett-Miller SM, Freeman B, Campbell GE, Dyer MA, Chen T, Stewart E, Koo S, Sheppard H, Easton J, Geeleher P. Bone morphogenetic protein (BMP) signaling determines neuroblastoma cell fate and sensitivity to retinoic acid. Nat Commun 2025; 16:2036. [PMID: 40021625 PMCID: PMC11871043 DOI: 10.1038/s41467-025-57185-y] [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: 07/03/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
Retinoic acid (RA) is a standard-of-care neuroblastoma drug thought to be effective by inducing differentiation. Curiously, RA has little effect on primary human tumors during upfront treatment but can eliminate neuroblastoma cells from the bone marrow during post-chemo maintenance therapy-a discrepancy that has never been explained. To investigate this, we treat a large cohort of neuroblastoma cell lines with RA and observe that the most RA-sensitive cells predominantly undergo apoptosis or senescence, rather than differentiation. We conduct genome-wide CRISPR knockout screens under RA treatment, which identify bone morphogenic protein (BMP) signaling as controlling the apoptosis/senescence vs differentiation cell fate decision and determining RA's overall potency. We then discover that BMP signaling activity is markedly higher in neuroblastoma patient samples at bone marrow metastatic sites, providing a plausible explanation for RA's ability to clear neuroblastoma cells specifically from the bone marrow, by seemingly mimicking interactions between BMP and RA during normal development.
Collapse
Affiliation(s)
- Min Pan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Yinwen Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - William C Wright
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xueying Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Barbara Passaia
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Duane Currier
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jonathan Low
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Richard H Chapple
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jacob A Steele
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jon P Connelly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Bensheng Ju
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Emily Plyler
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Meifen Lu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Allister J Loughran
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Burgess Freeman
- Preclinical Pharmacokinetic Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - George E Campbell
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Elizabeth Stewart
- Cellular Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Selene Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Paul Geeleher
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
6
|
Xu H, Zhang F, Xu Y, Chen T, Yuan F, Nie Q. Neuropathiazol induces neuronal-like differentiation in neuroblastoma cells via upregulation of PEG5. Pediatr Res 2025:10.1038/s41390-025-03925-1. [PMID: 39972157 DOI: 10.1038/s41390-025-03925-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/18/2024] [Accepted: 01/18/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Differentiation therapy is emerging as a promising strategy for treating neuroblastoma. However, the effects of neuropathiazol, a small molecule known to induce neuronal differentiation, have not been explored in neuroblastoma. PROCEDURE Neuroblastoma cell lines were used to investigate the effects of neuropathiazol and retinoic acid on cell morphology, proliferation, and invasion in vitro. In vivo, neuroblastoma cells were implanted in nude mice to assess neuropathiazol's therapeutic potential. Silver staining and markers of mature neurons were employed to evaluate neuropathiazol's ability to promote neuronal differentiation. RESULTS Neuropathiazol significantly inhibited the proliferation and invasion of neuroblastoma cells in vitro. It also enhanced synaptic growth and increased the expression of mature neuron markers more effectively than retinoic acid. Neuropathiazol treatment upregulated PEG5 expression, suggesting its role in promoting neuronal differentiation. Silencing PEG5 reversed these differentiation effects, reducing neuronal features. In vivo, neuropathiazol suppressed tumor growth and induced neuron-like differentiation in tumor tissues. However, its efficacy was diminished when PEG5 was knocked down. Additionally, neuropathiazol synergized with cyclophosphamide, enhancing its anti-neuroblastoma effects. CONCLUSION Neuropathiazol induces neuroblastoma differentiation, partly through PEG5 upregulation. As a promising differentiating agent for neuroblastoma, the combination of neuropathiazol and cyclophosphamide offers a potential treatment strategy for the disease. IMPACT Neuropathiazol significantly inhibits neuroblastoma cell proliferation and invasion in vitro. Neuropathiazol promotes synaptic growth and upregulates mature neuronal marker expression more effectively than retinoic acid. Neuropathiazol induces significant neuronal-like differentiation of neuroblastoma cells in vivo, leading to tumor growth suppression. PEG5 is identified as a critical mediator of neuropathiazol's differentiation-inducing effects. Knockdown of PEG5 reverses these effects, underscoring its pivotal role. The combination of neuropathiazol with cyclophosphamide synergistically enhances anti-neuroblastoma effects, offering a compelling pharmacotherapeutic strategy.
Collapse
Affiliation(s)
- Hao Xu
- Department of Pediatrics, Wuhan NO.1 Hospital, Wuhan, 430022, China
| | - Fei Zhang
- Department of Pediatrics, Wuhan Yaxin General Hospital, Wuhan, 430056, China
| | - Yi Xu
- Department of Endoscopy Center, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang, 330029, China
| | - Tianpeng Chen
- Department of Pharmacy, Fengcheng People's Hospital, Fengcheng, 331104, China
| | - Fenqian Yuan
- Department of Head and Neck Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, 330029, Jiangxi, China
| | - Qihong Nie
- Department of Oncology, Ganzhou People's Hospital, Ganzhou, 341000, China.
| |
Collapse
|
7
|
Amoroso L, Fagioli F, Tondo A, Conte M, Badino C, Di Cataldo A, Viscardi E, De Leonardis F, De Ioris MA, Luksch R. Strategies to manage the adverse effects of immunotherapy with dinutuximab beta in neuroblastoma: an Italian experience and literature review. Support Care Cancer 2025; 33:153. [PMID: 39907793 DOI: 10.1007/s00520-024-09135-5] [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: 03/19/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025]
Abstract
PURPOSE Patients with high-risk neuroblastoma (HR NB) frequently present with metastases in the bone marrow and bone. Approximately 15% of these patients are refractory to induction therapy, and 50% relapse. Dinutuximab beta is an anti-GD2 monoclonal antibody approved in Europe for maintenance therapy of pediatric patients with HR NB. Immunotherapy with anti-GD2 antibodies improves the survival of children with HR NB and relapsed or refractory disease. It is associated with adverse events, such as pain, fever, allergic reactions, capillary leak syndrome, and diarrhea. This manuscript aims to propose a practical guide in support and prevention treatment of adverse events. METHODS MEDLINE was searched using the terms "GD2," "ch14.18/CHO," "anti-GD2 antibody," "dinutuximab beta," and "neuroblastoma." The experts reappraised available evidence discussing different clinical Italian experiences. RESULTS Neuropathic pain is the main toxicity associated with dinutuximab beta and can be prevented with analgesics, including intravenous opioids and gabapentin by mouth. The intensity of the supportive treatment decreases from course to course. CONCLUSION In the experience of the authors, adverse events associated with dinutuximab beta may be prevented and managed in experienced centers. The supportive therapy may be reduced after the first cycle to improve the quality of life.
Collapse
Affiliation(s)
- Loredana Amoroso
- UOC Oncology, IRCCS Istituto G. Gaslini, UOC Oncology, Genova, Italy.
- Pediatric Oncology and Oncohematology Unit, Department of Maternal Infantile and Urological Sciences, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy.
| | - Franca Fagioli
- Pediatric Oncology Department, Regina Margherita Children's HospitalA.O.U. Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Annalisa Tondo
- Centro Di Eccellenza Di Oncologia Ed Ematologia Pediatrica, SOC "Oncologia, Ematologia, TCSE E Terapia Genica" IRCCS, AUO A. Meyer, Florence, Italy
| | - Massimo Conte
- UOC Oncology, IRCCS Istituto G. Gaslini, UOC Oncology, Genova, Italy
| | - Clara Badino
- UOC Oncology, IRCCS Istituto G. Gaslini, UOC Oncology, Genova, Italy
| | - Andrea Di Cataldo
- Department of Clinical and Experimental Medicine Unit of Pediatric Hematology and Oncology, Policlinico Hospital University of Catania, Catania, Italy
| | - Elisabetta Viscardi
- UOC Oncoematologia Pediatrica, DIDAS Salute Della Donna E del Bambino Azienda Ospedale-Università Padova, Padova, Italy
| | - Francesco De Leonardis
- UOC Di Pediatria Ad Indirizzo Onco Ematologico. Azienda Ospedaliero Universitaria Policlinico Di Bari, Bari, Italy
| | - Maria Antonietta De Ioris
- Dipartimento Di Onco-Ematologia E Terapia Cellulare E Genica, Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Roberto Luksch
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milano, Italy
| |
Collapse
|
8
|
Olgun N, Arayici ME, Kızmazoglu D, Cecen RE. Assessment of Chemo-Immunotherapy Regimens in Patients with Refractory or Relapsed Neuroblastoma: A Systematic Review with Meta-Analysis of Critical Oncological Outcomes. J Clin Med 2025; 14:934. [PMID: 39941606 PMCID: PMC11818460 DOI: 10.3390/jcm14030934] [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: 01/11/2025] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Neuroblastoma is a highly aggressive pediatric cancer, particularly in children with refractory or relapsed disease, where survival outcomes remain poor despite advancements in treatment. Combining anti-GD2 antibodies, such as dinutixumab beta, dinutixumab, and naxitanab, with conventional chemotherapy has emerged as a promising approach to improve clinical outcomes in this high-risk population. This chemo-immunotherapy regimen meta-analysis aimed to investigate the efficacy of these combination regimens by analyzing objective response rate (ORR), overall survival (OS), and event-free survival (EFS) outcomes across multiple studies. Methods: A systematic review and meta-analysis were conducted following PRISMA guidelines. PubMed, Web of Science, and Scopus databases were searched, yielding studies comprising the related reports. Both randomized controlled trials and non-randomized studies were included. The primary outcome of interest was ORR, and the secondary outcome of interest was EFS. A random-effects model using the DerSimonian-Laird method and Knapp-Hartung-Sidik-Jonkman adjustments was employed to pool effect sizes, and heterogeneity was assessed using I2 statistics. Results: A total of ten reports from eight studies were deemed eligible and included in the meta-analysis. The pooled ORR across the studies was 0.45 (95% CI: 0.35-0.54, p < 0.001), indicating that approximately 45% of patients showed a favorable treatment response, with moderate heterogeneity (I2 = 52.78%). The pooled analysis showed an OS of 75% (95% CI: 53-96, p < 0.001), and the pooled EFS effect size was 0.59 (95% CI: 0.45-0.73, p < 0.001), despite substantial heterogeneity (I2 = 60.54%). Conclusions: anti-GD2 antibodies combined with conventional chemotherapy may significantly improve response rates and event-free survival in children with refractory or relapsed neuroblastoma. Future research should focus on identifying predictive biomarkers to tailor therapies to individual patients, enhancing both efficacy and safety in this vulnerable population.
Collapse
Affiliation(s)
- Nur Olgun
- Department of Pediatric Oncology, Institute of Oncology, Dokuz Eylul University, İzmir 35340, Turkey; (D.K.); (R.E.C.)
- Acıbadem Kent Hospital, Karsiyaka, İzmir 35630, Turkey
| | - Mehmet Emin Arayici
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylul University, İzmir 35340, Turkey;
- Department of Public Health, Faculty of Medicine, Dokuz Eylul University, İzmir 35340, Turkey
| | - Deniz Kızmazoglu
- Department of Pediatric Oncology, Institute of Oncology, Dokuz Eylul University, İzmir 35340, Turkey; (D.K.); (R.E.C.)
| | - Refik Emre Cecen
- Department of Pediatric Oncology, Institute of Oncology, Dokuz Eylul University, İzmir 35340, Turkey; (D.K.); (R.E.C.)
| |
Collapse
|
9
|
Sultan I, Alfaar AS, Sultan Y, Salman Z, Qaddoumi I. Trends in childhood cancer: Incidence and survival analysis over 45 years of SEER data. PLoS One 2025; 20:e0314592. [PMID: 39752445 PMCID: PMC11698462 DOI: 10.1371/journal.pone.0314592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/12/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND The SEER Registry contains U.S. cancer statistics. To assess trends in incidence and survival and the impact of demographic factors among pediatric patients with cancer, we assessed nearly 5 decades (1975-2019) of data. METHODS All patients below the age of 20 with histology-confirmed malignancy were studied. Kaplan-Meier survival curves were generated to evaluate survival trends across treatment periods and ICCC classes. JoinPoint analysis was conducted to identify changes in incidence and survival. RESULTS The incidence of childhood cancer increased from 14.23 cases per 100,000 children in 1975-1979 to 18.89 in 2010-2019, with an average annual percent change of 0.73. This rise was more pronounced in several cancers, including leukemias, lymphomas, brain tumors, hepatic tumors, and gonadal germ cell tumors. Age-adjusted cancer mortality decreased from 4.9 to 2.3 per 100,000. Cancer-related mortality was consistently higher in boys than in girls, and in Black children than in White children. Survival significantly improved, with 5- and 10-year survival rates rising from 63.1% to 85.2% and from 58.8% to 82.7%, respectively. Leukemias showed a substantial increase in 5-year survival from 48.2% ± 1.7% to 85.1% ± 0.4% in 2010-2019. Lymphomas also showed significant improvement, with survival increasing from 72.9% ± 1.7% to 94.2% ± 0.3%. Despite these improvements, the survival of CNS tumors, bone tumors, and sarcomas remained suboptimal, with 5-year survival estimates of approximately 60%. Our joinpoint analysis confirmed our findings but revealed an interesting increase in the incidence of lymphomas limited to the years between 2005 and 2014. CONCLUSION This research elucidates advancements in survival among pediatric patients with cancer. The results offer critical perspectives on pediatric oncology, highlighting the imperative for ongoing innovation in therapeutics. Although the increase in incidence may partially stem from enhanced diagnostic capabilities and more comprehensive registration processes, the underlying causes remain unclear.
Collapse
Affiliation(s)
- Iyad Sultan
- Department of Pediatrics and Artificial Intelligence and Data Innovation Office (AIDI), King Hussein Cancer Center, Amman, Jordan
| | - Ahmad S. Alfaar
- Department of Ophthalmology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yaseen Sultan
- Department of Biostatistics, University of Iowa, Iowa City, IA, United States of America
| | - Zeena Salman
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| | - Ibrahim Qaddoumi
- Departments of Global Pediatric Medicine and Oncology, St. Jude Children’s Research Hospital, Memphis, TN, United States of America
| |
Collapse
|
10
|
Nong J, Su C, Li C, Wang C, Li W, Li Y, Chen P, Li Y, Li Z, She X, Yuan Z, Liu S, Chen C, Liao Q, Luo Y, Shi B. Global, regional, and national epidemiology of childhood neuroblastoma (1990-2021): a statistical analysis of incidence, mortality, and DALYs. EClinicalMedicine 2025; 79:102964. [PMID: 39720601 PMCID: PMC11667623 DOI: 10.1016/j.eclinm.2024.102964] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 12/26/2024] Open
Abstract
Background Neuroblastoma is the most prevalent extracranial solid tumor in pediatric populations worldwide, representing 8-10% of childhood malignancies and contributing to approximately 15% of pediatric cancer-related fatalities. This study aims to report global trends in the incidence, mortality, and disability-adjusted life years (DALYs) of childhood neuroblastoma from 1990 to 2021. Methods The study utilized data from the Global Burden of Disease (GBD) database to analyze neuroblastoma incidence, mortality, and DALYs in children aged 0-14 years. Rates for incidence, mortality, and DALYs were calculated per 100,000 population, with 95% uncertainty intervals (UIs). Data from 204 countries and territories were stratified by age, sex, and location. Trends were assessed using Joinpoint regression models to compute the annual percent change (APC) and log-transformed linear regression models to calculate the estimated average annual percentage change (EAPC). Findings Globally, the incidence of neuroblastoma in children in 2021 was 5560 cases (95% UI, 3734.21-7560.03), with 1977 deaths (95% UI, 1445.04-2528.54), and 174,186.30 DALYs (95% UI, 127,104.64-223,265.92). From 1990 to 2021, the incidence increased by 30.26% (95% UI, -1.24% to 72.51%), mortality by 20.35% (95% UI, -12.44% to 63.30%), and DALYs by 20.08% (95% UI, -12.89% to 63.27%). The incidence rate rose from 0.25 (95% UI, 0.18-0.33) per 100,000 individuals in 1990 to 0.28 (95% UI, 0.19-0.38) per 100,000 individuals in 2021, an overall increase of 12.60% (95% UI, -14.62% to 49.12%). Among the five Sociodemographic Index (SDI) regions, the highest EAPCs were observed in the low-to-mid SDI regions for incidence (1.87%; 95% CI, 1.64%-2.10%), mortality (1.22%; 95% CI, 1.09%-1.34%), and DALYs (1.36%; 95% CI, 1.15%-1.57%). Regionally, Central Asia exhibited the fastest annual increase in incidence (EAPC = 2.76%; 95% CI, 2.18%-3.34%). At the national level, India had the highest number of neuroblastoma cases globally in 2021, with 685 cases (95% UI, 404.16-1007.67). Interpretation The global trends for incidence, mortality, and DALYs related to pediatric neuroblastoma initially increased and then decreased, although an overall increasing trend was observed. However, the burden of disease remains significant in low-, low-middle-, and middle-SDI regions. A comprehensive understanding of the epidemiology of neuroblastoma in children is crucial for enhancing disease prevention and control efforts. Funding This research was funded by the Guangxi Natural Science Foundation (Grant No. 2024GXNSFAA010420) and the Youth Science Foundation of Guangxi Medical University (Grant No. GXMUYSF202404).
Collapse
Affiliation(s)
- Jusen Nong
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Cheng Su
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Changhua Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Congjun Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Wei Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Yong Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Peng Chen
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Yanqiang Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Zihao Li
- Department of Thoracic Surgery, Liuzhou People's Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi Zhuang Autonomous Region, PR China
| | - Xinjin She
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Zuxin Yuan
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Sentian Liu
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Chao Chen
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Qian Liao
- Department of Epidemiology and Biostatistics, School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Yige Luo
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| | - Bo Shi
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, PR China
| |
Collapse
|
11
|
Ye J, Jiang H, Tiche S, He C, Liu J, Bian F, Jedoui M, Forgo B, Islam MT, Zhao M, Emengo P, He B, Li Y, Li A, Truong A, Ho J, Simmermaker C, Yang Y, Zhou MN, Hu Z, Svensson K, Cuthbertson D, Hazard F, Xing L, Shimada H, Chiu B. Restoring Mitochondrial Quantity and Quality to Reverse Warburg Effect and Drive Tumor Differentiation. RESEARCH SQUARE 2024:rs.3.rs-5494402. [PMID: 39711563 PMCID: PMC11661309 DOI: 10.21203/rs.3.rs-5494402/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Reduced mitochondrial quality and quantity in tumors is associated with dedifferentiation and increased malignancy. However, it remains unclear how to restore mitochondrial quantity and quality in tumors, and whether mitochondrial restoration can drive tumor differentiation. Our study shows that restoring mitochondrial function using retinoic acid (RA) to boost mitochondrial biogenesis and a mitochondrial uncoupler to enhance respiration synergistically drives neuroblastoma differentiation and inhibits proliferation. U-13C-glucose/glutamine isotope tracing revealed a metabolic shift from the pentose phosphate pathway to oxidative phosphorylation, accelerating the TCA cycle and switching substrate preference from glutamine to glucose. These effects were reversed by ETC inhibitors or in ρ0 cells lacking mtDNA, emphasizing the necessity of mitochondrial function for differentiation. Dietary RA and uncoupler treatment promoted tumor differentiation in an orthotopic neuroblastoma xenograft model, evidenced by neuropil production and Schwann cell recruitment. Single-cell RNA sequencing analysis of the orthotopic xenografts revealed that this strategy effectively eliminated the stem cell population, promoted differentiation, and increased mitochondrial gene signatures along the differentiation trajectory, which could potentially significantly improve patient outcomes. Collectively, our findings establish a mitochondria-centric therapeutic strategy for inducing tumor differentiation, suggesting that maintaining/driving differentiation in tumor requires not only ATP production but also continuous ATP consumption and sustained ETC activity.
Collapse
|
12
|
Azechi T, Fukaya Y, Nitani C, Hara J, Kawamoto H, Taguchi T, Yoshimura K, Sato A, Hattori N, Ushijima T, Kimura T. Population Pharmacokinetics of Tamibarotene in Pediatric and Young Adult Patients with Recurrent or Refractory Solid Tumors. Curr Oncol 2024; 31:7155-7164. [PMID: 39590158 PMCID: PMC11592880 DOI: 10.3390/curroncol31110527] [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: 10/09/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Tamibarotene is a synthetic retinoid that inhibits tumor cell proliferation and promotes differentiation. We previously reported on the safety and tolerability of tamibarotene in patients with recurrent or refractory solid tumors. Therefore, in this study, we aimed to evaluate the pharmacokinetic properties of tamibarotene and construct a precise pharmacokinetic model. We also conducted a non-compartmental analysis and population pharmacokinetic (popPK) analysis based on the results of a phase I study. Targeted pediatric and young adult patients with recurrent or refractory solid tumors were administered tamibarotene at doses of 4, 6, 8, 10, and 12 g/m2/day. Serum tamibarotene concentrations were evaluated after administration, and a popPK model was constructed for tamibarotene using Phoenix NLME. During model construction, we considered the influence of various parameters (weight, height, body surface area, and age) as covariates. Notably, 22 participants were included in this study, and 109 samples were analyzed. A two-compartment model incorporating lag time was selected as the base model. In the final model, the body surface area was included as a covariate for apparent total body clearance, the central compartment volume of distribution, and the peripheral compartment volume of distribution. Visual prediction checks and bootstrap analysis confirmed the validity and predictive accuracy of the final model as satisfactory.
Collapse
Affiliation(s)
- Takuya Azechi
- Department of Pharmacy, Juntendo University Hospital, Bunkyo-ku, Tokyo 113-8431, Japan; (T.A.)
- Faculty of Pharmacy, Juntendo University, Urayasu 279-0013, Chiba, Japan
| | - Yutaka Fukaya
- Department of Pharmacy, Juntendo University Hospital, Bunkyo-ku, Tokyo 113-8431, Japan; (T.A.)
| | - Chika Nitani
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka 534-0021, Osaka, Japan; (C.N.); (J.H.)
| | - Junichi Hara
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka 534-0021, Osaka, Japan; (C.N.); (J.H.)
| | - Hiroshi Kawamoto
- Department of Pediatric and Allergy, Fujimi Clinic, Ota-ku, Tokyo 143-0015, Japan
| | - Tomoaki Taguchi
- Fukuoka College of Health Sciences, Fukuoka 814-0193, Fukuoka, Japan
| | - Kenichi Yoshimura
- Department of Biostatistics and Health Data Science, Nagoya City University Graduate School of Medical Science, Nagoya 467-8601, Aichi, Japan
| | - Akihiro Sato
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa 277-8577, Chiba, Japan
| | - Naoko Hattori
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Shinagawa-ku, Tokyo 142-8501, Japan; (N.H.)
| | - Toshikazu Ushijima
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Shinagawa-ku, Tokyo 142-8501, Japan; (N.H.)
| | - Toshimi Kimura
- Department of Pharmacy, Juntendo University Hospital, Bunkyo-ku, Tokyo 113-8431, Japan; (T.A.)
- Faculty of Pharmacy, Juntendo University, Urayasu 279-0013, Chiba, Japan
| |
Collapse
|
13
|
Weichert-Leahey N, Zimmerman MW, Berezovskaya A, Look AT, Abraham BJ. Accurate Measurement of Cell Number-Normalized Differential Gene Expression in Cells Treated With Retinoic Acid. Bio Protoc 2024; 14:e5106. [PMID: 39525967 PMCID: PMC11543784 DOI: 10.21769/bioprotoc.5106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 09/08/2024] [Accepted: 09/15/2024] [Indexed: 11/16/2024] Open
Abstract
Genome-wide gene expression analysis is a commonly used method to quantitatively examine the transcriptional signature of any tissue or cell state. Standard bulk cell RNA sequencing (RNA-seq) quantifies RNAs in the cells of the tissue type of interest through massive parallel sequencing of cDNA synthesized from the cellular RNA. The subsequent analysis of global RNA expression and normalization of RNA expression levels between two or more samples generally assumes that cells from all samples produce equivalent amounts of RNA per cell. This assumption may be invalid in cells where MYC or MYCN expression levels are markedly different and thus, overall mRNA expression per cell is altered. Here, we describe an approach for RNA-seq analysis of MYCN-amplified neuroblastoma cells during treatment with retinoic acid, which causes dramatic downregulation of MYCN expression and induces growth arrest and differentiation of the cells. Our procedure employs spiked-in RNA standards added in ratio to the number of cells in each sample prior to RNA extraction. In the analysis of differential gene expression, the expression level of each gene is standardized to the spiked-in RNA standard to accurately assess gene expression levels per cell in conditions of high and low MYCN expression. Our protocol thus provides a step-by-step experimental approach for normalizing RNA-seq expression data on a per-cell-number basis, allowing accurate assessment of differential gene expression in cells expressing markedly different levels of MYC or MYCN. Key features • High levels of MYC and MYCN expression in cancer cells cause substantial increases in the levels of overall mRNA expression per cell. • RNA-seq using control RNAs spiked-in on a per-cell basis more accurately reflects global expression changes, when comparing cell populations with substantially different MYCN expression levels. • In MYCN-amplified neuroblastoma, retinoic acid dramatically decreases MYCN expression levels, resulting in large changes in overall RNA expression levels per cell.
Collapse
Affiliation(s)
- Nina Weichert-Leahey
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - Mark W. Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alla Berezovskaya
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - A. Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - Brian J. Abraham
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
14
|
Inuwa RF, Moss D, Quayle J, Swadi R. Epac activation reduces trans-endothelial migration of undifferentiated neuroblastoma cells and cellular differentiation with a CDK inhibitor further enhances Epac effect. PLoS One 2024; 19:e0304547. [PMID: 39495719 PMCID: PMC11534210 DOI: 10.1371/journal.pone.0304547] [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: 05/14/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024] Open
Abstract
Neuroblastoma (NB) is the most common solid extracranial neoplasm found in children and is derived from primitive sympathoadrenal neural precursor. The disease accounts for 15% of all cancer deaths in children. The mortality rate is high in patients presenting with a metastatic tumour even with extensive treatments. This signifies the need for further research towards the development of new additional therapies that can combat not only tumour growth but metastasis, especially amongst the high-risk groups. During metastasis, primary tumour cells become migratory and travel towards a capillary within the tumour. They then degrade the matrix surrounding the pericytes and endothelial cells traversing the endothelial barrier twice to establish a secondary. This led to the hypothesis that modulation of the endothelial cell junctional stability could have an influence on tumour metastasis. To test this hypothesis, agents that modulate endothelial permeability on NB cell line migration and invasion were assessed in vitro in a tissue culture model. The cAMP agonist and its antagonists were found to have no obvious effect on both SK-N-BE2C and SK-N-AS migration, invasion and proliferation. Next, NB cells were cocultured with HDMEC cells and live cell imaging was used to assess the effect of an Epac agonist on trans-endothelial cell migration of NB cells. Epac1 agonist remarkably reduced the trans-endothelial migration of both SK-N-BE2C and SK-N-AS cells. These results demonstrate that an Epac1 agonist may perhaps serve as an adjuvant to currently existing therapies for the high-risk NB patients.
Collapse
Affiliation(s)
- Rabiu Fage Inuwa
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Faculty of Basic Medical Sciences, Department of Anatomy, College of Health Sciences, Bayero University, Kano, Nigeria
| | - Diana Moss
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - John Quayle
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- ALESS/ALESA Program, Center for Global Communication Strategies, Center for Global Education, University of Tokyo, Bunkyō, Japan
| | - Rasha Swadi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
- Institute for Health Research, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
15
|
Miera-Maluenda M, Pérez-Torres M, Mañas A, Rubio-San-Simón A, Butjosa-Espín M, Ruiz-Duran P, Seoane JA, Moreno L, Segura MF. Advances in the approaches used to repurpose drugs for neuroblastoma. Expert Opin Drug Discov 2024; 19:1309-1319. [PMID: 39258785 DOI: 10.1080/17460441.2024.2402413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Neuroblastoma (NB) remains a challenging pediatric malignancy with limited treatment options, particularly for high-risk cases. Drug repurposing offers a convenient and cost-effective strategy for treating rare diseases like NB. Using existing drugs with known safety profiles accelerates the availability of new treatments, reduces development costs, and mitigates risks, offering hope for improved patient outcomes in challenging conditions. AREAS COVERED This review provides an overview of the advances in approaches used to repurpose drugs for NB therapy. The authors discuss strategies employed in drug repurposing, including computational and experimental methods, and rational drug design, highlighting key examples of repurposed drugs with promising clinical results. Additionally, the authors examine the challenges and opportunities associated with drug repurposing in NB and discuss future directions and potential areas for further research. EXPERT OPINION The fact that only one new drug has been approved in the last 30 years for the treatment of neuroblastoma plus a significant proportion of high-risk NB patients that remain uncurable, evidences the need for new fast and cost-effective alternatives. Drug repurposing may accelerate the treatment development process while reducing expenses and risks. This approach can swiftly bring effective NB therapies to market, enhancing survival rates and patient quality of life.
Collapse
Affiliation(s)
- Marta Miera-Maluenda
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Pérez-Torres
- Department of Pediatric Oncology and Hematology, Vall D'Hebron University Hospital, Barcelona, Spain
| | - Adriana Mañas
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alba Rubio-San-Simón
- Pediatric Oncology and Hematology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Maria Butjosa-Espín
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paula Ruiz-Duran
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Lucas Moreno
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pediatric Oncology and Hematology, Vall D'Hebron University Hospital, Barcelona, Spain
| | - Miguel F Segura
- Childhood Cancer and Blood Disorders Group, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Tirelli M, Bonfiglio F, Cantalupo S, Montella A, Avitabile M, Maiorino T, Diskin SJ, Iolascon A, Capasso M. Integrative genomic analyses identify neuroblastoma risk genes involved in neuronal differentiation. Hum Genet 2024; 143:1293-1309. [PMID: 39192051 PMCID: PMC11522082 DOI: 10.1007/s00439-024-02700-2] [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: 06/12/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Genome-Wide Association Studies (GWAS) have been decisive in elucidating the genetic predisposition of neuroblastoma (NB). The majority of genetic variants identified in GWAS are found in non-coding regions, suggesting that they can be causative of pathogenic dysregulations of gene expression. Nonetheless, pinpointing the potential causal genes within implicated genetic loci remains a major challenge. In this study, we integrated NB GWAS and expression Quantitative Trait Loci (eQTL) data from adrenal gland to identify candidate genes impacting NB susceptibility. We found that ZMYM1, CBL, GSKIP and WDR81 expression was dysregulated by NB predisposing variants. We further investigated the functional role of the identified genes through computational analysis of RNA sequencing (RNA-seq) data from single-cell and whole-tissue samples of NB, neural crest, and adrenal gland tissues, as well as through in vitro differentiation assays in NB cell cultures. Our results indicate that dysregulation of ZMYM1, CBL, GSKIP, WDR81 may lead to malignant transformation by affecting early and late stages of normal program of neuronal differentiation. Our findings enhance the understanding of how specific genes contribute to NB pathogenesis by highlighting their influence on neuronal differentiation and emphasizing the impact of genetic risk variants on the regulation of genes involved in critical biological processes.
Collapse
Affiliation(s)
- Matilde Tirelli
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Ferdinando Bonfiglio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Sueva Cantalupo
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Annalaura Montella
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | | | - Teresa Maiorino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Sharon J Diskin
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, 19104, Philadelphia, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, 19104, Philadelphia, USA
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
| | - Mario Capasso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy.
| |
Collapse
|
17
|
Wu H, Zhang G, Liu Z, Liu W, Wang X, Zhao Y. Enhanced anti-tumor activity mediated by combination chimeric antigen receptor T cells targeting GD2 and GPC2 in high-risk neuroblastoma. Cytotherapy 2024; 26:1308-1319. [PMID: 38904586 DOI: 10.1016/j.jcyt.2024.05.023] [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: 01/26/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND AIMS Chimeric antigen receptor T (CAR-T) cells targeting single antigens show limited activity against solid tumors due to poor T cell persistence, low efficiency infiltration, and exhaustion together with heterogeneous tumor-associated antigen (TAA) expression. This is also true in high-risk neuroblastoma (HRNB), a lethal pediatric extracranial malignancy. To overcome these obstacles, a combinational strategy using GD2-specific and GPC2-specific CAR-T cells was developed to improve immunotherapeutic efficacy. METHODS We individually developed GD2-specific and GPC2-specific CARs containing a selective domain (sCAR) which was a peptide of 10 amino acids derived from human nuclear autoantigen La/SS-B. These constructs allowed us to generate two different HRNB antigen-specific CAR-T cells with enhanced biological activity through stimulating sCAR-engrafted T cells via a selective domain-specific monoclonal antibody (SmAb). Binding affinity and stimulation of GD2- and GPC2-specific sCARs by SmAb were measured, and transient and persistent anti-tumor cytotoxicity of GD2sCAR-T and GPC2sCAR-T cells were quantified in neuroblastoma cell lines expressing different TAA levels. The anti-tumor pharmaceutical effects and cellular mechanisms mediated by single or combinational sCAR-T cells were evaluated in vitro and in vivo. RESULTS GD2- and GPC2-specific sCARs had antigen-specific binding affinity similar to their parental counterparts and were recognized by SmAb. SmAb-mediated stimulation selectively activated sCAR-T proliferation and increased central memory T cells in the final products. SmAb-stimulated sCAR-T cells had enhanced transient cytolytic activity, and combination therapy extended long-term anti-tumor activity in vitro through TNF-α and IL-15 release. Stimulated sCAR-T cells overcame heterogeneous antigen expression in HRNB, and the multi-TAA-targeting strategy was especially efficacious in vivo, inducing apoptosis through the caspase-3/PARP pathway and inhibiting the release of several tumor-promoting cytokines. CONCLUSIONS These data suggest that combined targeting of multiple TAAs is a promising strategy to overcome heterogenous antigen expression in solid tumors and extend CAR-T cell persistence for HRNB immunotherapy.
Collapse
Affiliation(s)
- Huantong Wu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Guangji Zhang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Zhongfeng Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Weihua Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
| | - Xuan Wang
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Yu Zhao
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
18
|
Ren K, Wang Y, Zhang M, Tao T, Sun Z. Unveiling Tumorigenesis Mechanisms and Drug Therapy in Neuroblastoma by Mass Spectrometry Based Proteomics. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1323. [PMID: 39594898 PMCID: PMC11593200 DOI: 10.3390/children11111323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024]
Abstract
Neuroblastoma (NB) is the most common type of extracranial solid tumors in children. Despite the advancements in treatment strategies over the past years, the overall survival rate in patients within the high-risk NB group remains less than 50%. Therefore, new treatment options are urgently needed for this group of patients. Compared with genomic aberrations, proteomic alterations are more dynamic and complex, as well as more directly related to pathological phenotypes and external perturbations such as environmental changes and drug treatments. This review focuses on specific examples of proteomics application in various fundamental aspects of NB research, including tumorigenesis, drug treatment, drug resistance, and highlights potential protein signatures and related signaling pathways with translational values for clinical practice. Moreover, emerging cutting-edge proteomic techniques, such as single cell and spatial proteomics, as well as mass spectrometry imaging, are discussed for their potentials to probe intratumor heterogeneity of NB.
Collapse
Affiliation(s)
- Keyi Ren
- Department of Surgical Oncology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Yu Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Minmin Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250118, China
| | - Ting Tao
- Department of Surgical Oncology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou 310052, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou 310052, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250118, China
| |
Collapse
|
19
|
Flaadt T, Jaki C, Maier CP, Amorelli G, Klingebiel T, Schlegel PG, Eyrich M, Greil J, Schulte JH, Bader P, Handgretinger R, Lang P. Immune reconstitution after transplantation of autologous peripheral stem cells in children: a comparison between CD34+ selected and nonmanipulated grafts. Cytotherapy 2024; 26:1227-1235. [PMID: 38904583 DOI: 10.1016/j.jcyt.2024.05.013] [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: 11/29/2023] [Revised: 05/02/2024] [Accepted: 05/10/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND AND AIMS High-dose chemotherapy (HDC) followed by autologous stem cell transplantation (ASCT) improves the prognosis in pediatric patients with several solid tumors and lymphomas. Little is known about the reconstitution of the immune system after ASCT and the influence of CD34+ cell selection on the reconstitution in pediatric patients. METHODS Between 1990 and 2001, 94 pediatric patients with solid tumors and lymphomas received autologous CD34+ selected or unmanipulated peripheral stem cells after HDC. CD34+ selection was carried out with magnetic microbeads. The absolute numbers of T cells, B cells and natural killer (NK) cells were measured and compared in both groups at various time points post-transplant. RESULTS Recovery of T cells was significantly faster in the unmanipulated group at day 30, with no significant difference later on. Reconstitution of B and NK cells was similar in both groups without significant differences at any time. The CD34+-selected group was divided into patients receiving less or more than 5.385 × 106/kg CD34+ cells. Patients in the CD34+ high-dose group displayed significantly faster reconstitutions of neutrophiles and lymphocyte subsets than the CD34+ low-dose group. CONCLUSIONS Engraftment and reconstitution of leukocytes, B cells and NK cells after transplantation of CD34+ selected stem cells were comparable to that in patients receiving unmanipulated grafts. T-cell recovery was faster in the unmanipulated group only within the first month. However, this delay could be compensated by transplantation of >5.385 × 106 CD34+ cells/kg. Especially for patients receiving immunotherapy after HDC large numbers of immune effector cells such as NK and T cells are necessary to mediate antibody-dependent cellular cytotoxicity. Therefore, in patients receiving autologous CD34+-selected grafts, our data emphasize the need to administer high stem cell counts.
Collapse
Affiliation(s)
- Tim Flaadt
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany.
| | - Christina Jaki
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany; Simulation Center STUPS, Klinikum Stuttgart, Stuttgart, Germany
| | - Claus-Philipp Maier
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany; Department of Hematology, Oncology, Clinical Immunology and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Germano Amorelli
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Thomas Klingebiel
- Goethe University, University Hospital, Department of Pediatrics, Division for Stem Cell Transplantation and Immunology, Frankfurt, Germany
| | - Paul Gerhardt Schlegel
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University Medical Center, Wuerzburg, Germany
| | - Matthias Eyrich
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University Medical Center, Wuerzburg, Germany
| | - Johann Greil
- Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Johannes H Schulte
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Peter Bader
- Goethe University, University Hospital, Department of Pediatrics, Division for Stem Cell Transplantation and Immunology, Frankfurt, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Peter Lang
- Department of Hematology and Oncology, University Children's Hospital, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
20
|
Seo ES, Lee JW, Lim J, Shin S, Cho HW, Ju HY, Yoo KH, Sung KW, Park WY. Germline functional variants contribute to somatic mutation and outcomes in neuroblastoma. Nat Commun 2024; 15:8360. [PMID: 39333105 PMCID: PMC11437149 DOI: 10.1038/s41467-024-52128-5] [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: 06/19/2023] [Accepted: 08/27/2024] [Indexed: 09/29/2024] Open
Abstract
Germline genetic context may play a significant role in the development and evolution of cancer, particularly in childhood cancers such as neuroblastoma. This study investigates the role of putatively functional germline variants in neuroblastoma, even if they do not directly increase disease risk. Our whole-exome sequencing analysis of 125 patients with neuroblastoma reveals a positive correlation between germline variant burden and somatic mutations. Moreover, patients with higher germline variant burden exhibit worse outcomes. Similar findings are observed in the independent neuroblastoma cohort where a higher germline variant burden correlates with a higher somatic mutational burden and a worse overall survival outcome. However, contrasting results emerge in adult-onset cancer, emphasizing the importance of germline genetics in neuroblastoma. The enrichment of putatively functional germline variants in cancer predisposition genes is borderline significant when compared to healthy populations (P = 0.077; Odds Ratio, 1.45; 95% confidence intervals, 0.94-2.21) and significantly more pronounced against adult-onset cancers (P = 0.016; Odds Ratio, 2.13; 95% confidence intervals, 1.10-3.91). Additionally, the presence of these variants proves to have prognostic significance in neuroblastoma (log-rank P < 0.001), and combining germline with clinical risk factors notably improves survival predictions.
Collapse
Affiliation(s)
- Eun Seop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Department of Digital Health, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul, South Korea
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jinyeong Lim
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Sunghwan Shin
- Department of Laboratory Medicine, Inje University Ilsan Paik Hospital, Goyang, South Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| | - Woong-Yang Park
- Department of Digital Health, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul, South Korea.
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea.
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea.
| |
Collapse
|
21
|
Yang L, Huang K, Cao L, Ma Y, Li S, Zhou J, Zhao Z, Wang S. Molecular profiling of core immune-escape genes highlights TNFAIP3 as an immune-related prognostic biomarker in neuroblastoma. Inflamm Res 2024; 73:1529-1545. [PMID: 39028490 DOI: 10.1007/s00011-024-01914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most prevalent and deadliest pediatric solid tumor. With of over 50% of high-risk neuroblastoma cases relapse, the imperative for novel drug targets and therapeutic strategies is accentuated. In neuroblastoma, the existence of tumor-associated macrophages (TAMs) correlates with an unfavorable patient prognosis. However, the clinical relevance and prognostic implications of regulatory genes linked to TAMs infiltration in neuroblastoma remain unclear, and further study is required. METHODS We conducted a comprehensive analysis utilizing transcriptome expression profiles from three primary datasets associated with neuroblastoma (GSE45547, GSE49710, TARGET) to identify hub genes implicated in immune evasion within neuroblastoma. Subsequently, we utilized single-cell RNA sequencing analysis on 17 clinical neuroblastoma samples to investigate the expression and distribution of these hub genes, leading to the identification of TNFAIP3. The above three public databases were merged to allowed for the validation of TNFAIP3's molecular functions through GO and KEGG analysis. Furthermore, we assessed TNFAIP3's correlation with immune infiltration and its potential immunotherapeutic impact by multiple algorithms. Our single-cell transcriptome data revealed the role of TNFAIP3 in macrophage polarization. Finally, preliminary experimental verifications to confirm the biological functions of TNFAIP3-mediated TAMs in NB. RESULTS A total of 6 genes related to immune evasion were screened and we found that TNFAIP3 exhibited notably higher expression in macrophages than other immune cell types, based on the scRNA-sequencing data. GO and KEGG analysis showed that low expression of TNFAIP3 significantly correlated with the activation of multiple oncogenic pathways as well as immune-related pathways. Then validation affirmed that individuals within the TNFAIP3 high-expression cohort could potentially derive greater advantages from immunotherapeutic interventions, alongside exhibiting heightened immune responsiveness. Deciphering the pseudotime trajectory of macrophages, we revealed the potential of TNFAIP3 in inducing the polarization of macrophages towards the M1 phenotype. Finally, we confirmed that patients in the TNFAIP3 high expression group might benefit more from immunotherapy or chemotherapy as substantiated by RT-qPCR and immunofluorescence examinations. Moreover, the role of TNFAIP3 in macrophage polarization was validated. Preliminary experiment showed that TNFAIP3-mediated TAMs inhibit the proliferation, migration and invasion capabilities of NB cells. CONCLUSIONS Our results suggest that TNFAIP3 was first identified as a promising biomarker for immunotherapy and potential molecular target in NB. Besides, the presence of TNFAIP3 within TAMs may offer a novel therapeutic strategy for NB.
Collapse
Affiliation(s)
- Linyu Yang
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Huang
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lijian Cao
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Ma
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Suwen Li
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jianwu Zhou
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenzhen Zhao
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Shan Wang
- Department of Pediatric Surgical Oncology Children's Hospitial of Chongqinng Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
22
|
Sui X, Liu T, Zou Z, Zhang B, Zhang G. Effects and mechanisms of puerarin against neuroblastoma: insights from bioinformatics and in vitro experiments. BMC Complement Med Ther 2024; 24:257. [PMID: 38982456 PMCID: PMC11234716 DOI: 10.1186/s12906-024-04569-0] [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: 06/22/2023] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Neuroblastoma, a prevalent solid tumor in children, often manifests with hidden onset sites, rapid growth, and high metastatic potential. The prognosis for children with high-risk neuroblastoma remains poor, highlighting the urgent need for novel prognostic models and therapeutic avenues. In recent years, puerarin, as a kind of small molecule drug extracted from Chinese medicine Pueraria lobata, has demonstrated significant anticancer effects on various cancer cell types. In this study, through bioinformatics analysis and in vitro experiments, the potential and mechanism of puerarin in the treatment of neuroblastoma were investigated, and a prognostic model was established. METHODS A total of 9 drug-disease related targets were observed by constructing a database of drug targets and disease genes. Besides, GO and KEGG enrichment analysis was performed to explore the potential mechanism of its therapeutic effect. To construct the prognostic model, risk regression analysis and LASSO analysis were carried out for validation. Finally, the prognostic genes were identified. Parachute test and immunofluorescence staining were performed to verify the potential mechanism of puerarin in neuroblastoma treatment. RESULTS Three prognostic genes, i.e., BIRC5, TIMP2 and CASP9, were identified. In vitro studies verified puerarin's impact on BIRC5, TIMP2, and CASP9 expression, inhibiting proliferation in neuroblastoma SH-SY5Y cells. Puerarin disrupts the cytoskeleton, boosts gap junctional communication, curtailing invasion and migration, and induces mitochondrial damage in SH-SY5Y cells. CONCLUSIONS Based on network pharmacology and bioinformatics analysis, combined with in vitro experimental verification, puerarin was hereby observed to enhance GJIC in neuroblastoma, destroy cytoskeleton and thus inhibit cell invasion and migration, cause mitochondrial damage of tumor cells, and inhibit cell proliferation. Overall, puerarin, as a natural medicinal compound, does hold potential as a novel therapy for neuroblastoma.
Collapse
Affiliation(s)
- Xiaohui Sui
- Shandong University of Traditional Chinese Medicine, Jinan Shandong, 250014, China
| | - Tingting Liu
- Shandong University of Traditional Chinese Medicine, Jinan Shandong, 250014, China
| | - Zhiyun Zou
- Shandong University of Traditional Chinese Medicine, Jinan Shandong, 250014, China
| | - Baoqing Zhang
- Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan Shandong, 250011, China
| | - Guiju Zhang
- Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan Shandong, 250011, China.
| |
Collapse
|
23
|
Pathania AS, Chava H, Chaturvedi NK, Chava S, Byrareddy SN, Coulter DW, Challagundla KB. The miR-29 family facilitates the activation of NK-cell immune responses by targeting the B7-H3 immune checkpoint in neuroblastoma. Cell Death Dis 2024; 15:428. [PMID: 38890285 PMCID: PMC11189583 DOI: 10.1038/s41419-024-06791-7] [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: 11/20/2023] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
Neuroblastoma (NB) is a highly aggressive pediatric cancer that originates from immature nerve cells, presenting significant treatment challenges due to therapy resistance. Despite intensive treatment, approximately 50% of high-risk NB cases exhibit therapy resistance or experience relapse, resulting in poor outcomes often associated with tumor immune evasion. B7-H3 is an immune checkpoint protein known to inhibit immune responses. MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation. Our study aims to explore the impact of miRNAs on B7-H3 regulation, the anti-tumor immune response, and tumorigenicity in NB. Analysis of NB patients and patient-derived xenograft tumors revealed a correlation between higher B7-H3 expression and poorer patient survival. Notably, deceased patients exhibited a depletion of miR-29 family members (miR-29a, miR-29b, and miR-29c), which displayed an inverse association with B7-H3 expression in NB patients. Overexpression and knockdown experiments demonstrated that these miRNAs degrade B7-H3 mRNA, resulting in enhanced NK cell activation and cytotoxicity. In vivo, experiments provided further evidence that miR-29 family members reduce tumorigenicity, macrophage infiltration, and microvessel density, promote infiltration and activation of NK cells, and induce tumor cell apoptosis. These findings offer a rationale for developing more effective combination treatments that leverage miRNAs to target B7-H3 in NB patients.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Haritha Chava
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nagendra K Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Srinivas Chava
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Don W Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kishore B Challagundla
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- The Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
24
|
Amiri B, Yazdani Tabrizi M, Naziri M, Moradi F, Arzaghi M, Archin I, Behaein F, Bagheri Pour A, Ghannadikhosh P, Imanparvar S, Akhtari Kohneshahri A, Sanaye Abbasi A, Zerangian N, Alijanzadeh D, Ghayyem H, Azizinezhad A, Ahmadpour Youshanlui M, Poudineh M. Neuroprotective effects of flavonoids: endoplasmic reticulum as the target. Front Neurosci 2024; 18:1348151. [PMID: 38957188 PMCID: PMC11218733 DOI: 10.3389/fnins.2024.1348151] [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: 12/01/2023] [Accepted: 03/28/2024] [Indexed: 07/04/2024] Open
Abstract
The incidence of neurological disorders, particularly age-related neurodegenerative pathologies, exhibits an alarming upward trend, while current pharmacological interventions seldom achieve curative outcomes. Despite their diverse clinical presentations, neurological diseases often share a common pathological thread: the aberrant accumulation of misfolded proteins within the endoplasmic reticulum (ER). This phenomenon, known as ER stress, arises when the cell's intrinsic quality control mechanisms fail to cope with the protein-folding burden. Consequently, misfolded proteins accumulate in the ER lumen, triggering a cascade of cellular stress responses. Recognizing this challenge, researchers have intensified their efforts over the past two decades to explore natural compounds that could potentially slow or even reverse these devastating pathologies. Flavonoids constitute a vast and heterogeneous class of plant polyphenols, with over 10,000 identified from diverse natural sources such as wines, vegetables, medicinal plants, and organic products. Flavonoids are generally divided into six different subclasses: anthocyanidins, flavanones, flavones, flavonols, isoflavones, and flavonols. The diverse family of flavonoids, featuring a common phenolic ring backbone adorned with varying hydroxyl groups and additional modifications, exerts its antioxidant activity by inhibiting the formation of ROS, as evidenced by research. Also, studies suggest that polyphenols such as flavonoids can regulate ER stress through apoptosis and autophagy. By understanding these mechanisms, we can unlock the potential of flavonoids as novel therapeutic agents for neurodegenerative disorders. Therefore, this review critically examines the literature exploring the modulatory effects of flavonoids on various steps of the ER stress in neurological disorders.
Collapse
Affiliation(s)
- Bita Amiri
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Yazdani Tabrizi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdyieh Naziri
- Student Research Committee, School of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Moradi
- Student Research Committee, School of Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Arzaghi
- Department of Physical Education and Sports Science-Nutrition, Branch Islamic Azad University, Tehran, Iran
| | - Iman Archin
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Parna Ghannadikhosh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Imanparvar
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ata Akhtari Kohneshahri
- Student Research Committee, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Ali Sanaye Abbasi
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nasibeh Zerangian
- PhD Student in Health Education and Health Promotion, Department of Health Education and Health Promotion, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Dorsa Alijanzadeh
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hani Ghayyem
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arash Azizinezhad
- Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| | | | - Mohadeseh Poudineh
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
25
|
Friedman DN, Goodman PJ, Leisenring WM, Diller LR, Cohn SL, Howell RM, Smith SA, Tonorezos ES, Wolden SL, Neglia JP, Ness KK, Gibson TM, Nathan PC, Turcotte LM, Weil BR, Robison LL, Oeffinger KC, Armstrong GT, Sklar CA, Henderson TO. Impact of risk-based therapy on late morbidity and mortality in neuroblastoma survivors: a report from the Childhood Cancer Survivor Study. J Natl Cancer Inst 2024; 116:885-894. [PMID: 38460547 PMCID: PMC11160496 DOI: 10.1093/jnci/djae062] [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: 01/29/2024] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Early efforts at risk-adapted therapy for neuroblastoma are predicted to result in differential late effects; the magnitude of these differences has not been well described. METHODS Late mortality, subsequent malignant neoplasms (SMNs), and severe/life-threatening chronic health conditions (CHCs), graded according to CTCAE v4.03, were assessed among 5-year Childhood Cancer Survivor Study (CCSS) survivors of neuroblastoma diagnosed 1987-1999. Using age, stage at diagnosis, and treatment, survivors were classified into risk groups (low [n = 425]; intermediate [n = 252]; high [n = 245]). Standardized mortality ratios (SMRs) and standardized incidence ratios (SIRs) of SMNs were compared with matched population controls. Cox regression models estimated hazard ratios (HRs) and 95% confidence intervals for CHC compared with 1029 CCSS siblings. RESULTS Among survivors (49.8% male; median age = 21 years, range = 7-42; median follow-up = 19.3 years, range = 5-29.9), 80% with low-risk disease were treated with surgery alone, whereas 79.1% with high-risk disease received surgery, radiation, chemotherapy ± autologous stem cell transplant (ASCT). All-cause mortality was elevated across risk groups (SMRhigh = 27.7 [21.4-35.8]; SMRintermediate = 3.3 [1.7-6.5]; SMRlow = 2.8 [1.7-4.8]). SMN risk was increased among high- and intermediate-risk survivors (SIRhigh = 28.0 [18.5-42.3]; SIRintermediate = 3.7 [1.2-11.3]) but did not differ from the US population for survivors of low-risk disease. Compared with siblings, survivors had an increased risk of grade 3-5 CHCs, particularly among those with high-risk disease (HRhigh = 16.1 [11.2-23.2]; HRintermediate = 6.3 [3.8-10.5]; HRlow = 1.8 [1.1-3.1]). CONCLUSION Survivors of high-risk disease treated in the early days of risk stratification carry a markedly elevated burden of late recurrence, SMN, and organ-related multimorbidity, whereas survivors of low/intermediate-risk disease have a modest risk of late adverse outcomes.
Collapse
Affiliation(s)
- Danielle Novetsky Friedman
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| | - Pamela J Goodman
- Public Health Science Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Wendy M Leisenring
- Public Health Science Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lisa R Diller
- Department of Pediatrics, The Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan L Cohn
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Rebecca M Howell
- Division of Radiation Oncology, Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| | - Susan A Smith
- Division of Radiation Oncology, Department of Radiation Physics, The University of Texas at MD Anderson Cancer Center, Houston, TX, USA
| | - Emily S Tonorezos
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph P Neglia
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Todd M Gibson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Paul C Nathan
- Division of Haematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Lucie M Turcotte
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Brent R Weil
- Department of Pediatrics, The Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Surgery, Boston Children’s Hospital, Boston, MA, USA
| | - Leslie L Robison
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Kevin C Oeffinger
- Department of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Gregory T Armstrong
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, TN, USA
| | - Charles A Sklar
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pediatrics, Weill Cornell Medical College, New York, NY, USA
| | - Tara O Henderson
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
26
|
Choudhary S, Singh MK, Kashyap S, Seth R, Singh L. Wnt/β-Catenin Signaling Pathway in Pediatric Tumors: Implications for Diagnosis and Treatment. CHILDREN (BASEL, SWITZERLAND) 2024; 11:700. [PMID: 38929279 PMCID: PMC11201634 DOI: 10.3390/children11060700] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
The evolutionarily conserved Wnt signaling has a significant and diverse role in maintaining cell homeostasis and tissue maintenance. It is necessary in the regulation of crucial biological functions such as embryonal development, proliferation, differentiation, cell fate, and stem cell pluripotency. The deregulation of Wnt/β-catenin signaling often leads to various diseases, including cancer and non-cancer diseases. The role of Wnt/β-catenin signaling in adult tumors has been extensively studied in literature. Although the Wnt signaling pathway has been well explored and recognized to play a role in the initiation and progression of cancer, there is still a lack of understanding on how it affects pediatric tumors. This review discusses the recent developments of this signaling pathway in pediatric tumors. We also focus on understanding how different types of variations in Wnt signaling pathway contribute to cancer development and provide an insight of tissue specific mutations that lead to clinical progression of these tumors.
Collapse
Affiliation(s)
- Sahar Choudhary
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.C.); (R.S.)
| | | | - Seema Kashyap
- Department of Ocular Pathology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Rachna Seth
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.C.); (R.S.)
| | - Lata Singh
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi 110029, India; (S.C.); (R.S.)
| |
Collapse
|
27
|
Yoneda A, Shichino H, Hishiki T, Matsumoto K, Ohira M, Kamijo T, Kuroda T, Soejima T, Nakazawa A, Takimoto T, Yokota I, Teramukai S, Takahashi H, Fukushima T, Hara J, Kaneko M, Ikeda H, Tajiri T, Mugishima H, Nakagawara A. A nationwide phase II study of delayed local treatment for children with high-risk neuroblastoma: The Japan Children's Cancer Group Neuroblastoma Committee Trial JN-H-11. Pediatr Blood Cancer 2024; 71:e30976. [PMID: 38577760 DOI: 10.1002/pbc.30976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/15/2024] [Accepted: 02/13/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE Survival rates of patients with high-risk neuroblastoma are unacceptable. A time-intensified treatment strategy with delayed local treatment to control systemic diseases has been developed in Japan. We conducted a nationwide, prospective, single-arm clinical trial with delayed local treatment. This study evaluated the safety and efficacy of delayed surgery to increase treatment intensity. PATIENTS AND METHODS Seventy-five patients with high-risk neuroblastoma were enrolled in this study between May 2011 and September 2015. Delayed local treatment consisted of five courses of induction chemotherapy (cisplatin, pirarubicin, vincristine, and cyclophosphamide) and myeloablative high-dose chemotherapy (melphalan, etoposide, and carboplatin), followed by local tumor extirpation with surgery and irradiation. The primary endpoint was progression-free survival (PFS). The secondary endpoints were overall survival (OS), response rate, adverse events, and surgical complications. RESULTS Seventy-five patients were enrolled, and 64 were evaluable (stage 3, n = 8; stage 4, n = 56). The estimated 3-year PFS and OS rates (95% confidence interval [CI]) were 44.4% [31.8%-56.3%] and 80.7% [68.5%-88.5%], resspectively. The response rate of INRC after completion of the treatment protocol was 66% (42/64; 95% CI: 53%-77%; 23 CR [complete response], 10 VGPR [very good partial response], and nine PR [partial response]). None of the patients died during the protocol treatment or within 30 days of completion. Grade 4 adverse effects, excluding hematological adverse effects, occurred in 48% of patients [31/64; 95% CI: 36%-61%]. Major Surgical complications were observed in 25% of patients [13/51; 95% CI: 14%-40%]. CONCLUSION This study indicates that delayed local treatment is feasible and shows promising efficacy, suggesting that this treatment should be considered further in a comparative study of high-risk neuroblastoma.
Collapse
Affiliation(s)
- Akihiro Yoneda
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Surgery, Surgical Oncology, Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
- Pediatric Surgical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Shichino
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Pediatrics, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tomoro Hishiki
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Pediatric Surgery, Chiba University, Chiba, Japan
| | - Kimikazu Matsumoto
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Miki Ohira
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Takehiko Kamijo
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Tatsuo Kuroda
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Pediatric Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Toshinori Soejima
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Kobe Proton Center, Kobe, Japan
| | - Atsuko Nakazawa
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Clinical Research, Saitama Children's Medical Center, Saitama, Japan
| | - Tetsuya Takimoto
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Clinical Epidemiology Research Center for Pediatric Cancer, National Center for Child Health and Development, Tokyo, Japan
| | - Isao Yokota
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Biostatistics, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Satoshi Teramukai
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Biostatistics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideto Takahashi
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- National Institute of Public Health, Saitama, Japan
| | - Takashi Fukushima
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Department of Pediatric Hematology and Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Junichi Hara
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Michio Kaneko
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Ibaraki Prefectural Association of Health Evaluation and Promotion, Mito, Japan
| | - Hitoshi Ikeda
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Pediatric Surgery, Dokkyo Medical University Koshigaya Hospital, Koshigaya, Japan
| | - Tatsuro Tajiri
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideo Mugishima
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- Booth Memorial Aged Care Center GRACE, Tokyo, Japan
| | - Akira Nakagawara
- The Japan Children's Cancer Group (JCCG) Neuroblastoma Committee (JNBSG), Nagoya, Japan
- SAGA Heavy Ion Medical Accelerator in Tosu, Tosu, Japan
| |
Collapse
|
28
|
Pan M, Zhang Y, Wright WC, Liu X, Passaia B, Currier D, Low J, Chapple RH, Steele JA, Connelly JP, Lu M, Lee HM, Loughran AJ, Yang L, Abraham BJ, Pruett-Miller SM, Freeman B, Campbell GE, Dyer MA, Chen T, Stewart E, Koo S, Sheppard H, Easton J, Geeleher P. Bone morphogenetic protein (BMP) signaling determines neuroblastoma cell fate and sensitivity to retinoic acid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593394. [PMID: 38798584 PMCID: PMC11118433 DOI: 10.1101/2024.05.09.593394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Retinoic acid (RA) is a standard-of-care neuroblastoma drug thought to be effective by inducing differentiation. Curiously, RA has little effect on primary human tumors during upfront treatment but can eliminate neuroblastoma cells from the bone marrow during post-chemo consolidation therapy-a discrepancy that has never been explained. To investigate this, we treated a large cohort of neuroblastoma cell lines with RA and observed that the most RA-sensitive cells predominantly undergo apoptosis or senescence, rather than differentiation. We conducted genome-wide CRISPR knockout screens under RA treatment, which identified BMP signaling as controlling the apoptosis/senescence vs differentiation cell fate decision and determining RA's overall potency. We then discovered that BMP signaling activity is markedly higher in neuroblastoma patient samples at bone marrow metastatic sites, providing a plausible explanation for RA's ability to clear neuroblastoma cells specifically from the bone marrow, seemingly mimicking interactions between BMP and RA during normal development.
Collapse
|
29
|
Prete A, Lanino E, Saglio F, Biffi A, Calore E, Faraci M, Rondelli R, Favre C, Zecca M, Casazza G, Porta F, Luksch R, Cesaro S, Rabusin M, Parasole R, Mura RM, Lo Nigro L, Leardini D, Pagliara D, Locatelli F, Fagioli F. Phase II Study of Allogeneic Hematopoietic Stem Cell Transplantation for Children with High-Risk Neuroblastoma Using a Reduced-Intensity Conditioning Regimen: Results from the AIEOP Trial. Transplant Cell Ther 2024; 30:530.e1-530.e8. [PMID: 38460729 DOI: 10.1016/j.jtct.2024.03.002] [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: 09/08/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/11/2024]
Abstract
Despite aggressive multimodal treatment, the outcomes of pediatric patients with high-risk (HR) neuroblastoma (NB) remain poor. The rationale for allogeneic hematopoietic stem cell transplantation (allo-HCT) to treat NB was based on the possible graft-versus-tumor effect; however, toxicity limits its efficacy. We sought to prospectively assess the feasibility and efficacy of allo-HCT using a reduced-intensity conditioning regimen in pediatric patients with HR NB in a multicenter phase II trial. Primary endpoints were the rate of neutrophil and platelet engraftment, 5-year transplantation-related mortality (TRM), and disease-free survival (DFS). Secondary endpoint measures included the incidence of acute graft-versus-host disease (aGVHD) and chronic GVHD. Fifty-one patients were enrolled in the study. The 5-year cumulative incidence (CuI) of TRM was 29.4 ± 6.4%, and that of DFS was 11.8 ± 4.5%. Patients undergoing allo-HCT within 1 year of diagnosis or with bone marrow as their stem cell source had a higher DFS probability. The CuI of neutrophil engraftment, platelet engraftment, and grade II-IV aGVHD was 97.9 ± 2.1%, 93.8 ± 3.5%, and 47.1 ± 7.0%, respectively. The development of new therapeutic strategies could further improve disease control.
Collapse
Affiliation(s)
- Arcangelo Prete
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Edoardo Lanino
- Hematopoietic Stem Cell Transplantation Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Francesco Saglio
- Pediatric Oncohematology, Stem Cell Transplantation and Cell Therapy Division, AOU Città della Salute e della Scienza-Regina Margherita Children's Hospital, Turin, Italy
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, University-Hospital of Padua, Padua, Italy
| | - Elisabetta Calore
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, University-Hospital of Padua, Padua, Italy
| | - Maura Faraci
- Hematopoietic Stem Cell Transplantation Unit, IRCSS Istituto Giannina Gaslini, Genoa, Italy
| | - Roberto Rondelli
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Claudio Favre
- Department of Pediatric Hematology/Oncology and Hematopoietic Stem Cell Transplantation, Meyer Children's University Hospital, Florence, Italy
| | - Marco Zecca
- Department of Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gabriella Casazza
- Pediatric Oncohematology, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Fulvio Porta
- Pediatric Oncohematology and Bone Marrow Transplant Unit, Children's Hospital, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Roberto Luksch
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Simone Cesaro
- Pediatric Hematology Oncology Unit, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Marco Rabusin
- Department of Pediatrics, Institute of Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Rosanna Parasole
- Department of Pediatric Hemato-Oncology and Cellular Therapy, Azienda Sanitaria di Rilievo Nazionale Santobono-Pausilipon, Napoli, Italy
| | - Rosa Maria Mura
- Pediatric Oncology Unit, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Luca Lo Nigro
- Regional Reference Center for Pediatric Hematology and Oncology, Azienda Policlinico "G. Rodolico-San Marco", Catania, Italy
| | - Davide Leardini
- Pediatric Hematology and Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Daria Pagliara
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy
| | - Franca Fagioli
- Pediatric Oncohematology, Stem Cell Transplantation and Cell Therapy Division, AOU Città della Salute e della Scienza-Regina Margherita Children's Hospital, Turin, Italy; University of Turin, Turin, Italy
| |
Collapse
|
30
|
Munekane M, Fuchigami T, Ogawa K. Recent advances in the development of 225Ac- and 211At-labeled radioligands for radiotheranostics. ANAL SCI 2024; 40:803-826. [PMID: 38564087 PMCID: PMC11035452 DOI: 10.1007/s44211-024-00514-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 04/04/2024]
Abstract
Radiotheranostics utilizes a set of radioligands incorporating diagnostic or therapeutic radionuclides to achieve both diagnosis and therapy. Imaging probes using diagnostic radionuclides have been used for systemic cancer imaging. Integration of therapeutic radionuclides into the imaging probes serves as potent agents for radionuclide therapy. Among them, targeted alpha therapy (TAT) is a promising next-generation cancer therapy. The α-particles emitted by the radioligands used in TAT result in a high linear energy transfer over a short range, inducing substantial damage to nearby cells surrounding the binding site. Therefore, the key to successful cancer treatment with minimal side effects by TAT depends on the selective delivery of radioligands to their targets. Recently, TAT agents targeting biomolecules highly expressed in various cancer cells, such as sodium/iodide symporter, norepinephrine transporter, somatostatin receptor, αvβ3 integrin, prostate-specific membrane antigen, fibroblast-activation protein, and human epidermal growth factor receptor 2 have been developed and have made remarkable progress toward clinical application. In this review, we focus on two radionuclides, 225Ac and 211At, which are expected to have a wide range of applications in TAT. We also introduce recent fundamental and clinical studies of radiopharmaceuticals labeled with these radionuclides.
Collapse
Affiliation(s)
- Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
31
|
Dos Santos IL, Mitchell M, Nogueira PAS, Lafita-Navarro MC, Perez-Castro L, Eriom J, Kilgore JA, Williams NS, Guo L, Xu L, Conacci-Sorrell M. Targeting of neuroblastoma cells through Kynurenine-AHR pathway inhibition. FEBS J 2024; 291:2172-2190. [PMID: 38431776 DOI: 10.1111/febs.17109] [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: 10/19/2023] [Revised: 01/13/2024] [Accepted: 02/21/2024] [Indexed: 03/05/2024]
Abstract
Neuroblastoma poses significant challenges in clinical management. Despite its relatively low incidence, this malignancy contributes disproportionately to cancer-related childhood mortality. Tailoring treatments based on risk stratification, including MYCN oncogene amplification, remains crucial, yet high-risk cases often confront therapeutic resistance and relapse. Here, we explore the aryl hydrocarbon receptor (AHR), a versatile transcription factor implicated in diverse physiological functions such as xenobiotic response, immune modulation, and cell growth. Despite its varying roles in malignancies, AHR's involvement in neuroblastoma remains elusive. Our study investigates the interplay between AHR and its ligand kynurenine (Kyn) in neuroblastoma cells. Kyn is generated from tryptophan (Trp) by the activity of the enzymes indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO2). We found that neuroblastoma cells displayed sensitivity to the TDO2 inhibitor 680C91, exposing potential vulnerabilities. Furthermore, combining TDO2 inhibition with retinoic acid or irinotecan (two chemotherapeutic agents used to treat neuroblastoma patients) revealed synergistic effects in select cell lines. Importantly, clinical correlation analysis using patient data established a link between elevated expression of Kyn-AHR pathway genes and adverse prognosis, particularly in older children. These findings underscore the significance of the Kyn-AHR pathway in neuroblastoma progression, emphasizing its potential role as a therapeutic target.
Collapse
MESH Headings
- Humans
- Kynurenine/metabolism
- Neuroblastoma/pathology
- Neuroblastoma/metabolism
- Neuroblastoma/genetics
- Neuroblastoma/drug therapy
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Aryl Hydrocarbon/genetics
- Receptors, Aryl Hydrocarbon/antagonists & inhibitors
- Cell Line, Tumor
- Tryptophan Oxygenase/metabolism
- Tryptophan Oxygenase/genetics
- Tryptophan Oxygenase/antagonists & inhibitors
- Tretinoin/pharmacology
- Signal Transduction/drug effects
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors
- Cell Proliferation/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
Collapse
Affiliation(s)
- Igor Lopes Dos Santos
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael Mitchell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Dell Medical School, Austin, TX, USA
| | - Pedro A S Nogueira
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - M Carmen Lafita-Navarro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lizbeth Perez-Castro
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joyane Eriom
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jessica A Kilgore
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Guo
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maralice Conacci-Sorrell
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
32
|
Cocchi S, Greco V, Sidarovich V, Vigna J, Broso F, Corallo D, Zasso J, Re A, Rosatti EF, Longhi S, Defant A, Ladu F, Sanna V, Adami V, D’Agostino VG, Sturlese M, Sechi M, Aveic S, Mancini I, Sighel D, Quattrone A. EGCG Disrupts the LIN28B/Let-7 Interaction and Reduces Neuroblastoma Aggressiveness. Int J Mol Sci 2024; 25:4795. [PMID: 38732012 PMCID: PMC11084668 DOI: 10.3390/ijms25094795] [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: 03/21/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroblastoma (NB) is the most commonly diagnosed extracranial solid tumor in children, accounting for 15% of all childhood cancer deaths. Although the 5-year survival rate of patients with a high-risk disease has increased in recent decades, NB remains a challenge in pediatric oncology, and the identification of novel potential therapeutic targets and agents is an urgent clinical need. The RNA-binding protein LIN28B has been identified as an oncogene in NB and is associated with a poor prognosis. Given that LIN28B acts by negatively regulating the biogenesis of the tumor suppressor let-7 miRNAs, we reasoned that selective interference with the LIN28B/let-7 miRNA interaction would increase let-7 miRNA levels, ultimately leading to reduced NB aggressiveness. Here, we selected (-)-epigallocatechin 3-gallate (EGCG) out of 4959 molecules screened as the molecule with the best inhibitory activity on LIN28B/let-7 miRNA interaction and showed that treatment with PLC/PLGA-PEG nanoparticles containing EGCG (EGCG-NPs) led to an increase in mature let-7 miRNAs and a consequent inhibition of NB cell growth. In addition, EGCG-NP pretreatment reduced the tumorigenic potential of NB cells in vivo. These experiments suggest that the LIN28B/let-7 miRNA axis is a good therapeutic target in NB and that EGCG, which can interfere with this interaction, deserves further preclinical evaluation.
Collapse
Affiliation(s)
- Simona Cocchi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Valentina Greco
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Viktoryia Sidarovich
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Jacopo Vigna
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
- Department of Physics, University of Trento, 38123 Trento, Italy; (A.D.)
| | - Francesca Broso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Diana Corallo
- Istituto di Ricerca Pediatrica Fondazione Città della Speranza, 35127 Padova, Italy
| | - Jacopo Zasso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Angela Re
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Emanuele Filiberto Rosatti
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Sara Longhi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Andrea Defant
- Department of Physics, University of Trento, 38123 Trento, Italy; (A.D.)
| | - Federico Ladu
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (F.L.); (M.S.)
| | | | - Valentina Adami
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Vito G. D’Agostino
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Mattia Sturlese
- Molecular Modeling Section, Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35127 Padova, Italy;
| | - Mario Sechi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (F.L.); (M.S.)
| | - Sanja Aveic
- Istituto di Ricerca Pediatrica Fondazione Città della Speranza, 35127 Padova, Italy
| | - Ines Mancini
- Department of Physics, University of Trento, 38123 Trento, Italy; (A.D.)
| | - Denise Sighel
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| | - Alessandro Quattrone
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (S.C.); (V.G.); (V.S.); (V.G.D.)
| |
Collapse
|
33
|
Battistella ME, Freire NH, Toson B, Dalmolin M, Fernandes MAC, Tassinari ID, Jaeger M, Brunetto AT, Brunetto AL, Gregianin L, de Farias CB, Roesler R. Stemness and Cell Cycle Regulators and Their Modulation by Retinoic Acid in Ewing Sarcoma. Curr Issues Mol Biol 2024; 46:3990-4003. [PMID: 38785514 PMCID: PMC11119684 DOI: 10.3390/cimb46050246] [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: 03/11/2024] [Revised: 04/13/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Retinoic acid (RA) regulates stemness and differentiation in human embryonic stem cells (ESCs). Ewing sarcoma (ES) is a pediatric tumor that may arise from the abnormal development of ESCs. Here we show that RA impairs the viability of SK-ES-1 ES cells and affects the cell cycle. Cells treated with RA showed increased levels of p21 and its encoding gene, CDKN1A. RA reduced mRNA and protein levels of SRY-box transcription factor 2 (SOX2) as well as mRNA levels of beta III Tubulin (TUBB3), whereas the levels of CD99 increased. Exposure to RA reduced the capability of SK-ES-1 to form tumorspheres with high expression of SOX2 and Nestin. Gene expression of CD99 and CDKN1A was reduced in ES tumors compared to non-tumoral tissue, whereas transcript levels of SOX2 were significantly higher in tumors. For NES and TUBB3, differences between tumors and control tissue did not reach statistical significance. Low expression of CD99 and NES, and high expression of SOX2, were significantly associated with a poorer patient prognosis indicated by shorter overall survival (OS). Our results indicate that RA may display rather complex modulatory effects on multiple target genes associated with the maintenance of stem cell's features versus their differentiation, cell cycle regulation, and patient prognosis in ES.
Collapse
Affiliation(s)
- Maria Eduarda Battistella
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Natália Hogetop Freire
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Children’s Cancer Institute, Porto Alegre 90620-110, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
| | - Bruno Toson
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Matheus Dalmolin
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
- InovAI Lab, nPITI/IMD, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Marcelo A. C. Fernandes
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
- InovAI Lab, nPITI/IMD, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
- Department of Computer Engineering and Automation, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Isadora D. Tassinari
- Laboratory of Neurobiology and Metabolism (NeuroMet), Department of Physiology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Graduate Program in Physiology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Mariane Jaeger
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Children’s Cancer Institute, Porto Alegre 90620-110, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
| | - André T. Brunetto
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Children’s Cancer Institute, Porto Alegre 90620-110, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
| | - Algemir L. Brunetto
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Children’s Cancer Institute, Porto Alegre 90620-110, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
| | - Lauro Gregianin
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
- Department of Pediatrics, School of Medicine, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Pediatric Oncology Service, Clinical Hospital, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| | - Caroline Brunetto de Farias
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- Children’s Cancer Institute, Porto Alegre 90620-110, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
| | - Rafael Roesler
- Cancer and Neurobiology Laboratory, Experimental Research Center, Clinical Hospital (CPE-HCPA), Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
- National Science and Technology Institute for Children’s Cancer Biology and Pediatric Oncology—INCT BioOncoPed, Porto Alegre 90035-003, Brazil
- Department of Pharmacology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, Brazil
| |
Collapse
|
34
|
Alqathami MS, Khan MA, Yoosuf ABM. Global research trends in Total Body Irradiation: a bibliometric analysis. Front Oncol 2024; 14:1370059. [PMID: 38737901 PMCID: PMC11082912 DOI: 10.3389/fonc.2024.1370059] [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: 01/13/2024] [Accepted: 04/01/2024] [Indexed: 05/14/2024] Open
Abstract
Objectives This manuscript presents a bibliometric and visualization analysis of Total Body Irradiation (TBI) research, aiming to elucidate trends, gaps, and future directions in the field. This study aims to provide a comprehensive overview of the global research landscape of TBI, highlighting its key contributions, evolving trends, and potential areas for future exploration. Methods The data for this study were extracted from the Web of Science Core Collection (WoSCC), encompassing articles published up to May 2023. The analysis included original studies, abstracts, and review articles focusing on TBI-related research. Bibliometric indicators such as total publications (TP), total citations (TC), and citations per publication (C/P) were utilized to assess the research output and impact. Visualization tools such as VOS Viewer were employed for thematic mapping and to illustrate international collaboration networks. Results The analysis revealed a substantial body of literature, with 7,315 articles published by 2,650 institutions involving, 13,979 authors. Full-length articles were predominant, highlighting their central role in the dissemination of TBI research. The authorship pattern indicated a diverse range of scholarly influences, with both established and emerging researchers contributing significantly. The USA led in global contributions, with significant international collaborations observed. Recent research trends have focused on refining TBI treatment techniques, investigating long-term patient effects, and advancing dosimetry and biomarker studies for radiation exposure assessments. Conclusions TBI research exhibits a dynamic and multifaceted landscape, driven by global collaboration and innovation. It highlights the clinical challenges of TBI, such as its adverse effects and the need for tailored treatments in pediatric cases. Crucially, the study also acknowledges the fundamental science underpinning TBI, including its effects on inflammatory and apoptotic pathways, DNA damage, and the varied sensitivity of cells and tissues. This dual focus enhances our understanding of TBI, guiding future research toward innovative solutions and comprehensive care.
Collapse
Affiliation(s)
- Mamdouh Saud Alqathami
- Department of Oncology, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | - Ahamed Badusha Mohamed Yoosuf
- Department of Oncology, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
35
|
Minò A, Lopez F, Barbaro R, Barile M, Ambrosone L, Colella M. Effects of Anionic Liposome Delivery of All- Trans-Retinoic Acid on Neuroblastoma Cell Differentiation. Biomimetics (Basel) 2024; 9:257. [PMID: 38786467 PMCID: PMC11118614 DOI: 10.3390/biomimetics9050257] [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: 03/29/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
All-trans-retinoic acid (ATRA) has long been known to affect cell growth and differentiation. To improve ATRA's therapeutic efficacy and pharmacodynamics, several delivery systems have been used. In this study, free ATRA and anionic-liposome-encapsulated ATRA were compared for their effects on SK-N-SH human neuroblastoma cell growth and differentiation. Anionic liposomes made of L-α-phosphatidylcholine (PC) and L-α-phosphatidic acid (PA), empty (PC-PA) and loaded with ATRA (PC-PA-ATRA), were characterized by dynamic light scattering (DLS) and electrophoretic mobility measurements, and drug entrapment efficiency (EE%) was measured to evaluate the applicability of the new colloidal formulation. The results of brightfield microscopy and cell growth curves indicated that ATRA, whether free or encapsulated, reduced growth and induced differentiation, resulting in SK-N-SH cells changing from epithelioid to neuronal-like morphologies, and producing a significant increase in neurite growth. To further characterize the neuro-differentiation of SK-N-SH cells, the expression of βIII-Tubulin and synaptophysin and mitochondria localization were analyzed via immunofluorescence. Increased expression of neuronal markers and a peculiar localization of mitochondria in the neuritic extensions were apparent both in ATRA- and PC-PA-ATRA-differentiated cells. As a whole, our results strongly indicate that ATRA treatment, by any means, can induce the differentiation of parent SK-N-SH, and they highlight that its encapsulation in anionic liposomes increases its differentiation ability in terms of the percentage of neurite-bearing cells. Interestingly, our data also suggest an unexpected differentiation capability of anionic liposomes per se. This work highlights the importance of developing and carefully testing novel delivery nanocarriers, which are a necessary first "step" in the development of new therapeutic settings.
Collapse
Affiliation(s)
- Antonio Minò
- Department of Biosciences and Territory (DiBT), University of Molise, Contrada Lappone, 86090 Pesche, Italy;
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Via F. De Sanctis snc, 86100 Campobasso, Italy;
| | - Francesco Lopez
- Department of Agricultural, Environmental and Food Sciences (DiAAA), University of Molise, Via F. De Sanctis snc, 86100 Campobasso, Italy;
| | - Roberto Barbaro
- Department of Biosciences, Biotechnology and Environment (DBBA), University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy; (R.B.); (M.B.)
| | - Maria Barile
- Department of Biosciences, Biotechnology and Environment (DBBA), University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy; (R.B.); (M.B.)
| | - Luigi Ambrosone
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Via F. De Sanctis snc, 86100 Campobasso, Italy;
| | - Matilde Colella
- Department of Biosciences, Biotechnology and Environment (DBBA), University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy; (R.B.); (M.B.)
| |
Collapse
|
36
|
Takenouchi A, Kudo W, Terui K, Komatsu S, Oita S, Yoshizawa H, Kawaguchi Y, Fumita T, Nishimura K, Hishiki T. Impaired Aortic Growth in Neuroblastoma Patients After Intensive Treatment. J Pediatr Surg 2024; 59:593-598. [PMID: 38184434 DOI: 10.1016/j.jpedsurg.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 01/08/2024]
Abstract
PURPOSE This study evaluated the abdominal aortic diameter in high-risk neuroblastoma (NB) patients and the risk of aortic narrowing following intensive treatment. METHODS We measured the aortic diameter at four specific levels of the abdominal aorta (diaphragmatic crus, celiac axis, and the root of the superior (SMA) and inferior (IMA) mesenteric arteries) on contrast CT scans. The control group consisted of 56 children with non-oncologic disorders, while the NB group included 35 patients with high-risk abdominal NB. We used regression analysis of age and aortic diameter to determine the regression formula for each level in each group and performed intergroup comparisons using t-test. RESULTS We evaluated a total of 160 contrast-enhanced CTs performed in the 35 eligible cases. The aortic diameter of pretreated NB patients was not significantly different from the controls. After receiving any treatment, the aortic diameter was significantly smaller in the NB group (p < 0.01 each). Patients who underwent radical surgery, particularly gross total resection (n = 26), had smaller aortic diameters at all levels compared to controls (p < 0.01 each). Patients treated with radiotherapy (RT) had smaller aortic diameters than controls. External beam radiotherapy (EBRT) patients (n = 24) had smaller aortic diameters at all levels except the celiac axis (crus, SMA, IMA; p < 0.01 each), and intraoperative radiotherapy (IORT) ± EBRT patients (n = 5) had smaller aortic diameters at all levels (p < 0.01 each). CONCLUSION Patients with NB may experience impaired development of the abdominal aorta after multimodal therapy, particularly after RT. Close observation and long-term follow-up is essential to monitor for catastrophic vascular complications. LEVEL OF EVIDENCE LEVEL III.
Collapse
Affiliation(s)
- Ayako Takenouchi
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Wataru Kudo
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keita Terui
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shugo Komatsu
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoru Oita
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroko Yoshizawa
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yunosuke Kawaguchi
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takashi Fumita
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Katsuhiro Nishimura
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoro Hishiki
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
37
|
Żebrowska U, Balwierz W, Wechowski J, Wieczorek A. Survival Benefit of Myeloablative Therapy with Autologous Stem Cell Transplantation in High-Risk Neuroblastoma: A Systematic Literature Review. Target Oncol 2024; 19:143-159. [PMID: 38401028 DOI: 10.1007/s11523-024-01033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Multimodal treatment of newly diagnosed high-risk neuroblastoma (HRNB) includes induction chemotherapy, consolidation with myeloablative therapy (MAT) and autologous stem cell transplantation (ASCT), followed by anti-disialoganglioside 2 (GD2) immunotherapy, as recommended by the Children's Oncology Group (COG) and the Society of Paediatric Oncology European Neuroblastoma (SIOPEN). Some centres proposed an alternative approach with induction chemotherapy followed by anti-GD2 immunotherapy, without MAT+ASCT. OBJECTIVE The aim of this systematic literature review was to compare survival outcomes in patients with HRNB treated with or without MAT+ASCT and with or without subsequent anti-GD2 immunotherapy. PATIENTS AND METHODS The review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. MEDLINE via PubMed and EMBASE databases were systematically searched for randomised controlled trials (RCT) and observational comparative studies in patients with HRNB using search terms for 'neuroblastoma' and ('myeloablative therapy' OR 'stem cell transplantation'). Reporting of at least one survival outcome [event-free survival (EFS), progression-free survival, relapse-free survival and/or overall survival (OS)] was required for inclusion. Outcomes from RCTs were synthesized in meta-analysis, while meta-analysis of non-RCTs was not planned owing to expected heterogeneity. RESULTS Literature searches produced 2587 results with 41 publications reporting 34 comparative studies included in the review. Of these, 7 publications reported 4 RCTs, and 34 publications reported 30 non-RCT studies. Studies differed with respect to included populations, induction regimen, response to induction, additional treatments and transplantation procedures. Subsequent treatments of relapse were rarely reported and could not be compared. In the meta-analysis, EFS was in favour of MAT+ASCT over conventional chemotherapy or no further treatment [hazard ratio (HR) = 0.78, 95% confidence interval (CI) 0.67-0.91, p = 0.001] with a trend favouring MAT+ASCT for OS (HR = 0.86, 95% CI 0.73-1.00, p = 0.05). Tandem MAT+ASCT was found to improve EFS compared with the single procedure, with improvement in both EFS and OS in patients treated with anti-GD2 therapy. Non-RCT comparative studies were broadly consistent with evidence from the RCTs; however, not all reported survival benefits of MAT+ASCT (single or tandem). Limited comparative evidence on treatment without MAT+ASCT in patients treated with anti-GD2 immunotherapy suggests an increased risk of relapse. In relapsed patients, MAT+ASCT appears to improve OS, but evidence remains scarce. CONCLUSIONS Survival benefits in patients treated with MAT+ASCT confirm that the procedure should remain an integral part of multimodal therapy. In patients treated with anti-GD2 immunotherapy, limited evidence suggests that omitting MAT+ASCT is associated with an increased risk of relapse, and therefore, a change in clinical practice can currently not be recommended. Evidence suggests the use of tandem MAT+ASCT compared with the single procedure, with greater benefits observed in patients treated with anti-GD2 immunotherapy. Limited evidence also suggests improved survival following MAT+ASCT in relapsed patients, which needs to be viewed in light of emerging chemoimmunotherapy in this setting.
Collapse
Affiliation(s)
- Urszula Żebrowska
- Department of Paediatric Oncology and Haematology, University Children's Hospital of Krakow, 265 Wielicka str, 30-663, Krakow, Poland
| | - Walentyna Balwierz
- Department of Paediatric Oncology and Haematology, University Children's Hospital of Krakow, 265 Wielicka str, 30-663, Krakow, Poland
- Department of Paediatric Oncology and Haematology, Jagiellonian University Medical College, 265 Wielicka str, 30-663, Krakow, Poland
| | - Jarosław Wechowski
- EUSA Pharma, Breakspear Park, Breakspear Way, Hemel Hempstead, HP2 4TZ, UK
| | - Aleksandra Wieczorek
- Department of Paediatric Oncology and Haematology, University Children's Hospital of Krakow, 265 Wielicka str, 30-663, Krakow, Poland.
- Department of Paediatric Oncology and Haematology, Jagiellonian University Medical College, 265 Wielicka str, 30-663, Krakow, Poland.
| |
Collapse
|
38
|
Cheng H, Lin Y, Yang W, Chang X, Feng J, Yang S, Liu S, Yu T, Peng X, Zheng P, Zhang C, Jia H, Qin H, Wang H. Visual conservation treatment dilemmas in neuroblastoma with bilateral blindness. Discov Oncol 2024; 15:48. [PMID: 38403682 PMCID: PMC10894793 DOI: 10.1007/s12672-024-00898-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 02/19/2024] [Indexed: 02/27/2024] Open
Abstract
OBJECTIVE To investigate the clinical features, treatment strategies, and prognosis of neuroblastoma with bilateral blindness. METHODS The clinical data of five patients with bilateral blindness neuroblastoma admitted to Beijing Children's Hospital from April 2018 to September 2020 were retrospectively collected to summarize their clinical characteristics. RESULTS All patients were female and the median age at presentation was 25 (23, 41) months. The median intervention time from the onset of symptoms of bilateral blindness to the start of treatment was 10 (10, 12) days. All five cases were staged as stage M and grouped as high risk. Four cases were MYCN gene amplification and one case was MYCN acquisition. Five children were treated according to a high-risk neuroblastoma treatment protocol. Four children did not recover their vision after treatment, and one case improved to have light perception. All patients were effectively followed up for a median of 20 (12, 31) months, with three deaths, one tumor-free survival, and one recurrent tumor-bearing survival. CONCLUSION Neuroblastoma with bilateral blindness is rare in the clinic, mostly in children of young age, and is often associated with MYCN amplification and multiple metastases. Early hormone shock therapy and optic nerve decompression are beneficial for preserving the child's vision. A joint multi-disciplinary treatment may help in the formulation of treatment decisions. Achieving a balance between good visual preservation and survival within the short optic nerve neurotherapeutic window is extremely challenging.
Collapse
Affiliation(s)
- Haiyan Cheng
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Yu Lin
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Wei Yang
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Xiaofeng Chang
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Jun Feng
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Shen Yang
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Shan Liu
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Tong Yu
- Medical Imaging Center, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Xiaojiao Peng
- Department of Neurosurgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Panpan Zheng
- Department of Ophthalmology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Chengyue Zhang
- Department of Ophthalmology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Haiwei Jia
- Department of Radiotherapy, Beijing Fengtai You Anmen Hospital, Beijing, 10069, China
| | - Hong Qin
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Huanmin Wang
- Department of Oncology Surgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
- MOE Key Laboratory of Major Diseases in Children, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
| |
Collapse
|
39
|
Makimoto A, Fujisaki H, Matsumoto K, Takahashi Y, Cho Y, Morikawa Y, Yuza Y, Tajiri T, Iehara T. Retinoid Therapy for Neuroblastoma: Historical Overview, Regulatory Challenges, and Prospects. Cancers (Basel) 2024; 16:544. [PMID: 38339295 PMCID: PMC10854948 DOI: 10.3390/cancers16030544] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Retinoids are vitamin A derivatives and include trans-retinoic acid, isotretinoin, tamibarotene, and bexarotene, all of which are currently available for clinical use. The clinical development of retinoid therapy for neuroblastoma has a history spanning more than four decades. The most promising agent is isotretinoin, which can contribute to improving event-free survival in patients with high-risk neuroblastoma by approximately 10% when administered over six months as maintenance therapy. Although isotretinoin is regarded as an essential component in the standard clinical management of high-risk neuroblastoma, its use for this purpose in the US and EU is off-label. To promote isotretinoin use in Japan as a treatment for neuroblastoma, our clinical research team is planning to launch an investigator-initiated, registration-directed clinical trial. The present review article discusses the basic science behind retinoid therapy, pre-clinical/clinical evidence on neuroblastoma, the concept of the proposed clinical trial, and prospects for this therapy.
Collapse
Affiliation(s)
- Atsushi Makimoto
- Department of Laboratory Medicine, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan
- Clinical Research Support Center, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
- Department of Hematology/Oncology, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
| | - Hiroyuki Fujisaki
- Department of Pediatric Hematology/Oncology, Osaka City General Hospital, Osaka 534-0021, Japan;
| | - Kimikazu Matsumoto
- Children’s Cancer Center, National Center for Child Health and Development, Tokyo 157-8535, Japan;
| | - Yoshiyuki Takahashi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan;
| | - Yuko Cho
- Department of Pediatrics, Hokkaido University Hospital, Sapporo 060-8648, Japan;
| | - Yoshihiko Morikawa
- Clinical Research Support Center, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
| | - Yuki Yuza
- Department of Hematology/Oncology, Tokyo Metropolitan Children’s Medical Center, Fuchu 183-8561, Japan;
| | - Tatsuro Tajiri
- Department of Pediatric Surgery, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan;
| | - Tomoko Iehara
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| |
Collapse
|
40
|
Jiang H, Tiche SJ, He CJ, Jedoui M, Forgo B, Zhao M, He B, Li Y, Li AM, Truong AT, Ho J, Simmermaker C, Yang Y, Zhou MN, Hu Z, Cuthbertson DJ, Svensson KJ, Hazard FK, Shimada H, Chiu B, Ye J. Mitochondrial uncoupler and retinoic acid synergistically induce differentiation and inhibit proliferation in neuroblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576741. [PMID: 38328117 PMCID: PMC10849550 DOI: 10.1101/2024.01.22.576741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Neuroblastoma is a leading cause of death in childhood cancer cases. Unlike adult malignancies, which typically develop from aged cells through accumulated damage and mutagenesis, neuroblastoma originates from neural crest cells with disrupted differentiation. This distinct feature provides novel therapeutic opportunities beyond conventional cytotoxic methods. Previously, we reported that the mitochondrial uncoupler NEN (niclosamide ethanolamine) activated mitochondria respiration to reprogram the epigenome, promoting neuronal differentiation. In the current study, we further combine NEN with retinoic acid (RA) to promote neural differentiation both in vitro and in vivo. The treatment increased the expression of RA signaling and neuron differentiation-related genes, resulting in a global shift in the transcriptome towards a more favorable prognosis. Overall, these results suggest that the combination of a mitochondrial uncoupler and the differentiation agent RA is a promising therapeutic strategy for neuroblastoma.
Collapse
Affiliation(s)
- Haowen Jiang
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Clifford JiaJun He
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Mohamed Jedoui
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Balint Forgo
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Meng Zhao
- Department of Pathology, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Bo He
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Yang Li
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Albert M. Li
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Jestine Ho
- Agilent Technologies, Inc., Santa Clara, CA, USA
| | | | - Yanan Yang
- Agilent Technologies, Inc., Santa Clara, CA, USA
| | - Meng-Ning Zhou
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Zhen Hu
- Olivia Consulting Service, Redwood City, CA, USA
| | | | - Katrin J. Svensson
- Department of Pathology, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Bill Chiu
- Department of Surgery, Stanford University, Stanford, CA, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
41
|
Lee JH, Jaiswal MS, Jang YS, Choi JH, Kim GC, Hong JW, Hwang DS. No-ozone cold plasma induces apoptosis in human neuroblastoma cell line via increased intracellular reactive oxygen species (ROS). BMC Complement Med Ther 2024; 24:46. [PMID: 38245726 PMCID: PMC10799363 DOI: 10.1186/s12906-023-04313-0] [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: 08/29/2022] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND This study aimed to evaluate the effect of argon-based No-ozone Cold Plasma (NCP) on neuroblastoma cancer cell apoptosis. METHODS Experiments were performed with SK-N-SH and HS 68. Cell cultures were treated with NCP for 1, 3, and 5 min. NCP was applied using three different strategies: direct NCP application to cell cultures, to only media, and to only cells. Evaluation of cell viability and the level of the reactive oxygen species (ROS) was performed. N-acetyl-L-cysteine (NAC) was also used to antagonize intracellular ROS. Cleaved caspase 3, PARP, aquaporin (AQP) 3 and 8 were detected. RESULTS NCP induced a gradual decrease in the SK-N-SH cell viability. In contrast, the viability of HS 68 cells did not change. SK-N-SH cells viability was reduced the most when the only media-NCP application strategy was employed. Intracellular ROS levels were significantly increased with time. Cleaved caspase 3 and PARP were increased at 6 h after NCP application. SK-N-SH cells remained viable with NAC after NCP application. AQP 3 and 8 were over-expressed in SK-N-SH cells. CONCLUSION These findings demonstrate the anti-cancer effect of NCP on neuroblastoma cells. NCP enhanced the selective apoptosis of neuroblastoma cells due to the increased intracellular ROS.
Collapse
Affiliation(s)
- Jung-Han Lee
- Department of Oral and Maxillofacial Surgery, Dental and Life Science Institute, Dental School, Pusan National University, Busan, South Korea
| | - M Shriya Jaiswal
- Department of Oral and Maxillofacial Surgery, Dental and Life Science Institute, Dental School, Pusan National University, Busan, South Korea
| | - Yoon-Seo Jang
- Department of Oral and Maxillofacial Surgery, Dental and Life Science Institute, Dental School, Pusan National University, Busan, South Korea
| | - Jeong-Hae Choi
- Department of Research and Development, FEAGLE Corporations, 70-6, Jeungsan-ro, Mulgeum-eup, Yangsan-si, 50614, Gyeongsangnam-do, South Korea
| | - Gyoo-Cheon Kim
- Department of Research and Development, FEAGLE Corporations, 70-6, Jeungsan-ro, Mulgeum-eup, Yangsan-si, 50614, Gyeongsangnam-do, South Korea
- Department of Oral Anatomy and Cell Biology, School of Dentistry, Pusan National University, Busan, South Korea
| | - Jin-Woo Hong
- Department of Internal Medicine, School of Korean Medicine, Yangsan Campus of Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, 50612, Gyeongsangnam-do, South Korea.
| | - Dae-Seok Hwang
- Department of Oral and Maxillofacial Surgery, Dental and Life Science Institute, Dental School, Pusan National University, Busan, South Korea.
- Dental Research Institute, Pusan National University Dental Hospital, Yangsan, South Korea.
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Pusan National University, Beomeo-ri, Mulgeum-eup, Yangsan-si, 50612, Gyeongsangnam-do, South Korea.
| |
Collapse
|
42
|
Cermakova K, Tao L, Dejmek M, Sala M, Montierth MD, Chan YS, Patel I, Chambers C, Loeza Cabrera M, Hoffman D, Parchem RJ, Wang W, Nencka R, Barbieri E, Hodges HC. Reactivation of the G1 enhancer landscape underlies core circuitry addiction to SWI/SNF. Nucleic Acids Res 2024; 52:4-21. [PMID: 37993417 PMCID: PMC10783513 DOI: 10.1093/nar/gkad1081] [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: 08/23/2023] [Revised: 09/29/2023] [Accepted: 10/27/2023] [Indexed: 11/24/2023] Open
Abstract
Several cancer core regulatory circuitries (CRCs) depend on the sustained generation of DNA accessibility by SWI/SNF chromatin remodelers. However, the window when SWI/SNF is acutely essential in these settings has not been identified. Here we used neuroblastoma (NB) cells to model and dissect the relationship between cell-cycle progression and SWI/SNF ATPase activity. We find that SWI/SNF inactivation impairs coordinated occupancy of non-pioneer CRC members at enhancers within 1 hour, rapidly breaking their autoregulation. By precisely timing inhibitor treatment following synchronization, we show that SWI/SNF is dispensable for survival in S and G2/M, but becomes acutely essential only during G1 phase. We furthermore developed a new approach to analyze the oscillating patterns of genome-wide DNA accessibility across the cell cycle, which revealed that SWI/SNF-dependent CRC binding sites are enriched at enhancers with peak accessibility during G1 phase, where they activate genes involved in cell-cycle progression. SWI/SNF inhibition strongly impairs G1-S transition and potentiates the ability of retinoids used clinically to induce cell-cycle exit. Similar cell-cycle effects in diverse SWI/SNF-addicted settings highlight G1-S transition as a common cause of SWI/SNF dependency. Our results illustrate that deeper knowledge of the temporal patterns of enhancer-related dependencies may aid the rational targeting of addicted cancers.
Collapse
Affiliation(s)
- Katerina Cermakova
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Ling Tao
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Milan Dejmek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michal Sala
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Matthew D Montierth
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA
| | - Yuen San Chan
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Ivanshi Patel
- Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | - Courtney Chambers
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Translational Biology and Molecular Medicine Graduate Program, Houston, TX, USA
| | - Mario Loeza Cabrera
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Dane Hoffman
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Ronald J Parchem
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Radim Nencka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eveline Barbieri
- Department of Pediatrics, Section of Hematology-Oncology, Texas Children's Cancer and Hematology Center, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - H Courtney Hodges
- Department of Molecular and Cellular Biology, and Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
| |
Collapse
|
43
|
Cherkaoui S, Yang L, McBride M, Turn CS, Lu W, Eigenmann C, Allen GE, Panasenko OO, Zhang L, Vu A, Liu K, Li Y, Gandhi OH, Surrey L, Wierer M, White E, Rabinowitz JD, Hogarty MD, Morscher RJ. Reprogramming neuroblastoma by diet-enhanced polyamine depletion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.573662. [PMID: 38260457 PMCID: PMC10802427 DOI: 10.1101/2024.01.07.573662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Neuroblastoma is a highly lethal childhood tumor derived from differentiation-arrested neural crest cells1,2. Like all cancers, its growth is fueled by metabolites obtained from either circulation or local biosynthesis3,4. Neuroblastomas depend on local polyamine biosynthesis, with the inhibitor difluoromethylornithine showing clinical activity5. Here we show that such inhibition can be augmented by dietary restriction of upstream amino acid substrates, leading to disruption of oncogenic protein translation, tumor differentiation, and profound survival gains in the TH-MYCN mouse model. Specifically, an arginine/proline-free diet decreases the polyamine precursor ornithine and augments tumor polyamine depletion by difluoromethylornithine. This polyamine depletion causes ribosome stalling, unexpectedly specifically at adenosine-ending codons. Such codons are selectively enriched in cell cycle genes and low in neuronal differentiation genes. Thus, impaired translation of these codons, induced by the diet-drug combination, favors a pro-differentiation proteome. These results suggest that the genes of specific cellular programs have evolved hallmark codon usage preferences that enable coherent translational rewiring in response to metabolic stresses, and that this process can be targeted to activate differentiation of pediatric cancers.
Collapse
Affiliation(s)
- Sarah Cherkaoui
- Pediatric Cancer Metabolism Laboratory, Children’s Research Center, University of Zurich, 8032 Zurich, Switzerland
- Division of Oncology, University Children’s Hospital Zurich and Children’s Research Center, University of Zurich, 8032 Zurich, Switzerland
| | - Lifeng Yang
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton University, Princeton, NJ 08544, USA
| | - Matthew McBride
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton University, Princeton, NJ 08544, USA
| | - Christina S. Turn
- Division of Oncology and Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton University, Princeton, NJ 08544, USA
| | - Caroline Eigenmann
- Pediatric Cancer Metabolism Laboratory, Children’s Research Center, University of Zurich, 8032 Zurich, Switzerland
- Division of Oncology, University Children’s Hospital Zurich and Children’s Research Center, University of Zurich, 8032 Zurich, Switzerland
| | - George E. Allen
- Bioinformatics Support Platform, Faculty of Medicine, University of Geneva 1211, Switzerland
- Department of Microbiology and Molecular Medicine, Institute of Genetics and Genomics Geneva, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Olesya O. Panasenko
- Department of Microbiology and Molecular Medicine, Institute of Genetics and Genomics Geneva, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- BioCode: RNA to proteins (R2P) Platform, University of Geneva, 1211 Geneva, Switzerland
| | - Lu Zhang
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08901, USA
- Department of Molecular Biology and Biochemistry, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Annette Vu
- Division of Oncology and Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kangning Liu
- Division of Oncology and Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yimei Li
- Division of Oncology and Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Om H. Gandhi
- Division of Oncology and Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lea Surrey
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Michael Wierer
- Proteomics Research Infrastructure, Panum Institute, Blegdamsvej 3B, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Eileen White
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08901, USA
- Department of Molecular Biology and Biochemistry, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Joshua D. Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton University, Princeton, NJ 08544, USA
| | - Michael D. Hogarty
- Division of Oncology and Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raphael J. Morscher
- Pediatric Cancer Metabolism Laboratory, Children’s Research Center, University of Zurich, 8032 Zurich, Switzerland
- Division of Oncology, University Children’s Hospital Zurich and Children’s Research Center, University of Zurich, 8032 Zurich, Switzerland
- Division of Human Genetics, Medical University Innsbruck, Peter-Mayr-Str. 1, 6020 Innsbruck, Austria
| |
Collapse
|
44
|
Kim H, Kim HJ, Jo Y, Yoon SH, Koh YK, Kang S, Koh KN, Im HJ. Ten-Year Trends of Hematopoietic Stem Cell Transplantation in Korean Pediatric Cancer from the National Health Insurance Claims Data. Cancer Res Treat 2024; 56:294-304. [PMID: 37680122 PMCID: PMC10789968 DOI: 10.4143/crt.2023.598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/03/2023] [Indexed: 09/09/2023] Open
Abstract
PURPOSE We aimed to determine the current application and survival trends of hematopoietic stem cell transplantation (HSCT) among Korean children and adolescents with cancer. MATERIALS AND METHODS Data of patients aged < 20 years with KCD-10 (Korean Classifications of Diseases, 10th revision) C codes and specific designation codes were collected from the National Health Insurance Service database. Thirty claim codes for HSCT were included, and data from 2009 to 2019 were analyzed. RESULTS The operational definition of pediatric cancer yielded an annual average of 2,000, with annual cases decreasing. In 2019, 221 HSCTs were performed, a decrease from the ten-year average of 276. Allografts outnumbered autografts with a ratio of 1.5:1. The source of allograft was bone marrow in 15% of patients in 2009; however, it substantially decreased to 3.3% in 2019. Furthermore, 70.5% of allogeneic HSCT used peripheral blood stem cell (PBSC) grafts, which increased to 89.3% by 2015. Cord blood utilization markedly decreased to 2.7% in 2018. The 5-year overall survival (OS) rate of all patients was 85.1%. Overall mortality decreased among patients who underwent recent HSCT, and they exhibited a higher 5-year OS rate. CONCLUSION In Korea, the number of pediatric patients with cancer is declining; however, the ratio of transplants to all patients remains constant. Patients who recently underwent transplantation showed better survival rates, possibly due to HSCT optimization. Korea showed a substantially greater PBSC utilization in pediatric HSCT. An in-depth examination encompassing donor relations and cause of death with a prospective registry is required in future studies.
Collapse
Affiliation(s)
- Hyery Kim
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hwa Jung Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Youngjun Jo
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Su Hyun Yoon
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Kwon Koh
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Pediatrics, Chosun University College of Medicine, Gwangju, Korea
| | - Sunghan Kang
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung-Nam Koh
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho Joon Im
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
45
|
Abstract
Neuroblastoma is a malignant tumor of neuroblasts, immature nerve cells found in several areas of the body. It usually affects children under age of 5. As usual, the tumor has ability to grow rapidly and to expand vastly which ultimately leads to death. Mostly, management decisions can be drawn by the prediction of the stage of the disease as well as age at the time of its diagnosis. There are four main stages of neuroblastoma, and treatment is according to the low and high risk of the disease. Several cytotoxic agents along with other therapies (antibody therapy, gene therapy, and even immunological therapies, antiangiogenic therapy, etc.) are used. Immunotherapy also has an important treatment option used nowadays for neuroblastoma. The discovery of major neuroblastoma-predisposition gene anaplastic lymphoma kinase cause somatic transformation or gene strengthening in diagnosed neuroblastoma. Promising new antiangiogenic strategies have also been introduced for the treatment of neuroblastoma with multiple mylomas. To manage numerous myelomas and cancers, including neuroblastoma, bone marrow transplantation and peripheral blood stem cell transplantation may be used.
Collapse
Affiliation(s)
- Muhammad Imran Qadir
- Institute of Molecular Biology & Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Bilal Ahmed
- University of Science And Technology of Fujairah, UAE; School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sumaira Noreen
- Faculty of Pharmaceutical Sciences, Governemnet College University, Faisalabad, Pakistan
| |
Collapse
|
46
|
Kushner BH, LaQuaglia MP, Cardenas FI, Basu EM, Gerstle JT, Kramer K, Roberts SS, Wolden SL, Cheung NKV, Modak S. Stage 4N neuroblastoma before and during the era of anti-G D2 immunotherapy. Int J Cancer 2023; 153:2019-2031. [PMID: 37602920 PMCID: PMC11925214 DOI: 10.1002/ijc.34693] [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: 06/06/2023] [Revised: 07/28/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023]
Abstract
Patients with stage 4N neuroblastoma (distant metastases limited to lymph nodes) stand out as virtually the only survivors of high-risk neuroblastoma (HR-NB) before myeloablative therapy (MAT) and immunotherapy with anti-GD2 monoclonal antibodies (mAbs) became standard. Because no report presents more recent results with 4N, we analyzed our large 4N experience. All 51 pediatric 4N patients (<18 years old) diagnosed 1985 to 2021 were reviewed. HR-NB included MYCN-nonamplified 4N diagnosed at age ≥18 months and MYCN-amplified 4N. Among 34 MYCN-nonamplified high-risk patients, 20 are relapse-free 1.5+ to 37.5+ (median 12.5+) years post-diagnosis, including 13 without prior MAT and 5 treated with little (1 cycle; n = 2) or no mAb (n = 3), while 14 patients (7 post-MAT, 8 post-mAbs) relapsed (all soft tissue). Of 15 MYCN-amplified 4N patients, 7 are relapse-free 2.1+ to 26.4+ (median 11.6+) years from the start of chemotherapy (all received mAbs; 3 underwent MAT) and 4 are in second remission 4.2+ to 21.8+ years postrelapse (all soft tissue). Statistical analyses showed no significant association of survival with either MAT or mAbs for MYCN-nonamplified HR-NB; small numbers prevented these analyses for MYCN-amplified patients. The two patients with intermediate-risk 4N (14-months-old) are relapse-free 7+ years postresection of primary tumors; distant disease spontaneously regressed. The natural history of 4N is marked by NB confined to soft tissue without early relapse in bones or bone marrow, where mAbs have proven efficacy. These findings plus curability without MAT, as seen elsewhere and at our center, support consideration of treatment reduction for MYCN-nonamplified 4N.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael P LaQuaglia
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Justin T Gerstle
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Suzanne L Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
47
|
Chennakesavalu M, Pudela C, Applebaum MA, Lee SM, Che Y, Naranjo A, Park JR, Volchenboum SL, Henderson TO, Cohn SL, Desai AV. Persistence of Racial and Ethnic Disparities in Risk and Survival for Patients with Neuroblastoma over Two Decades. EJC PAEDIATRIC ONCOLOGY 2023; 2:100022. [PMID: 38213818 PMCID: PMC10783478 DOI: 10.1016/j.ejcped.2023.100022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
BACKGROUND Racial/ethnic survival disparities in neuroblastoma were first reported more than a decade ago. We sought to investigate if these disparities have persisted with current era therapy. METHODS Two patient cohorts were identified in the International Neuroblastoma Risk Group Data Commons (INRGdc) (Cohort 1: diagnosed 2001-2009, n=4359; Cohort 2: diagnosed 2010-2019, n=4891). Chi-squared tests were used to assess the relationship between race/ethnicity and clinical and biologic features. Survival was estimated by the Kaplan-Meier method. Cox proportional hazards regression analyses were performed to investigate the association between racial/ethnic groups and prognostic markers. RESULTS Significantly higher 5-year event-free survival (EFS) and overall survival (OS) were observed for Cohort 2 compared to Cohort 1 (P<0.001 and P<0.001, respectively). Compared to White patients, Black patients in both cohorts had a higher proportion of high-risk disease (Cohort 1: P<0.001; Cohort 2: P<0.001) and worse EFS (Cohort 1: P<0.001; Cohort 2 P<0.001) and OS (Cohort 1: P<0.001; Cohort 2: P<0.001). In Cohort 1, Native Americans also had a higher proportion of high-risk disease (P=0.03) and inferior EFS/OS. No significant survival disparities were observed for low- or intermediate-risk patients in either cohort or high-risk patients in Cohort 1. Hispanic patients with high-risk disease in Cohort 2 had significantly inferior OS (P=0.047). Significantly worse OS, but not EFS, (P=0.006 and P=0.02, respectively) was also observed among Black and Hispanic patients assigned to receive post-Consolidation dinutuximab on clinical trials (n=885). CONCLUSION Racial/ethnic survival disparities have persisted over time and were observed among high-risk patients assigned to receive post-Consolidation dinutuximab.
Collapse
Affiliation(s)
| | - Caileigh Pudela
- MedStar Georgetown University Hospital, Washington, D.C., USA
| | | | - Sang Mee Lee
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Yan Che
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Arlene Naranjo
- Children’s Oncology Group Statistics and Data Center, Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Julie R. Park
- St. Jude Children’s Research Hospital, Memphis, TN, USA
| | | | | | - Susan L. Cohn
- Department of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Ami V. Desai
- Department of Pediatrics, University of Chicago, Chicago, IL, USA
| |
Collapse
|
48
|
Guyer RA, Picard N, Mueller JL, Ohishi K, Leavitt A, Murphy AJ, Cornejo KM, Hotta R, Goldstein AM. Differentiated neuroblastoma cells remain epigenetically poised for de-differentiation to an immature state. Dis Model Mech 2023; 16:dmm049754. [PMID: 38095019 PMCID: PMC10810560 DOI: 10.1242/dmm.049754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/20/2023] [Indexed: 12/28/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor of childhood and accounts for a significant share of childhood cancer deaths. Prior studies utilizing RNA sequencing of bulk tumor populations showed two predominant cell states characterized by high and low expression of neuronal genes. Although cells respond to treatment by altering their gene expression, it is unclear whether this reflects shifting balances of distinct subpopulations or plasticity of individual cells. Using mouse and human neuroblastoma cell lines lacking MYCN amplification, we show that the antigen CD49b (also known as ITGA2) distinguishes these subpopulations. CD49b expression marked proliferative cells with an immature gene expression program, whereas CD49b-negative cells expressed differentiated neuronal marker genes and were non-cycling. Sorted populations spontaneously switched between CD49b expression states in culture, and CD49b-negative cells could generate rapidly growing, CD49b-positive tumors in mice. Although treatment with the chemotherapy drug doxorubicin selectively killed CD49b-positive cells in culture, the CD49b-positive population recovered when treatment was withdrawn. We profiled histone 3 (H3) lysine 27 acetylation (H3K27ac) to identify enhancers and super enhancers that were specifically active in each population and found that CD49b-negative cells maintained the priming H3 lysine 4 methylation (H3K4me1) mark at elements that were active in cells with high expression of CD49b. Improper maintenance of primed enhancer elements might thus underlie cellular plasticity in neuroblastoma, representing potential therapeutic targets for this lethal tumor.
Collapse
Affiliation(s)
- Richard A. Guyer
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nicole Picard
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jessica L. Mueller
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Kensuke Ohishi
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co. Ltd., Akitakata, Hiroshima 739-1195, Japan
| | - Abigail Leavitt
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Andrew J. Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN 38015, USA
| | - Kristine M. Cornejo
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
49
|
Chaudhry KA, Jacobi JJ, Gillard BM, Karasik E, Martin JC, da Silva Fernandes T, Hurley E, Feltri ML, Attwood KM, Twist CJ, Smiraglia DJ, Long MD, Bianchi-Smiraglia A. Aryl hydrocarbon receptor is a tumor promoter in MYCN-amplified neuroblastoma cells through suppression of differentiation. iScience 2023; 26:108303. [PMID: 38026169 PMCID: PMC10654598 DOI: 10.1016/j.isci.2023.108303] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/25/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in children. MYCN amplification is detected in almost half of high-risk cases and is associated with poorly differentiated tumors, poor patient prognosis and poor response to therapy, including retinoids. We identify the aryl hydrocarbon receptor (AhR) as a transcription factor promoting the growth and suppressing the differentiation of MYCN-amplified neuroblastoma. A neuroblastoma specific AhR transcriptional signature reveals an inverse correlation of AhR activity with patients' outcome, suggesting AhR activity is critical for disease progression. AhR modulates chromatin structures, reducing accessibility to regions responsive to retinoic acid. Genetic and pharmacological inhibition of AhR results in induction of differentiation. Importantly, AhR antagonism with clofazimine synergizes with retinoic acid in inducing differentiation both in vitro and in vivo. Thus, we propose AhR as a target for MYCN-amplified neuroblastoma and that its antagonism, combined with current standard-of-care, may result in a more durable response in patients.
Collapse
Affiliation(s)
- Kanita A. Chaudhry
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Justine J. Jacobi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Bryan M. Gillard
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ellen Karasik
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jeffrey C. Martin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Edward Hurley
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
| | - Maria Laura Feltri
- Department of Biochemistry and Neurology, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY, USA
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Foundation I.R.C.C.S. Carlo Besta Neurological Institute Milan, Italy
| | - Kristopher M. Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Clare J. Twist
- Department of Pediatric Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Dominic J. Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mark D. Long
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Anna Bianchi-Smiraglia
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
50
|
Searcy MB, Larsen RK, Stevens BT, Zhang Y, Jin H, Drummond CJ, Langdon CG, Gadek KE, Vuong K, Reed KB, Garcia MR, Xu B, Kimbrough DW, Adkins GE, Djekidel N, Porter SN, Schreiner PA, Pruett-Miller SM, Abraham BJ, Rehg JE, Hatley ME. PAX3-FOXO1 dictates myogenic reprogramming and rhabdomyosarcoma identity in endothelial progenitors. Nat Commun 2023; 14:7291. [PMID: 37968277 PMCID: PMC10651858 DOI: 10.1038/s41467-023-43044-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 10/27/2023] [Indexed: 11/17/2023] Open
Abstract
Fusion-positive rhabdomyosarcoma (FP-RMS) driven by the expression of the PAX3-FOXO1 (P3F) fusion oncoprotein is an aggressive subtype of pediatric rhabdomyosarcoma. FP-RMS histologically resembles developing muscle yet occurs throughout the body in areas devoid of skeletal muscle highlighting that FP-RMS is not derived from an exclusively myogenic cell of origin. Here we demonstrate that P3F reprograms mouse and human endothelial progenitors to FP-RMS. We show that P3F expression in aP2-Cre expressing cells reprograms endothelial progenitors to functional myogenic stem cells capable of regenerating injured muscle fibers. Further, we describe a FP-RMS mouse model driven by P3F expression and Cdkn2a loss in endothelial cells. Additionally, we show that P3F expression in TP53-null human iPSCs blocks endothelial-directed differentiation and guides cells to become myogenic cells that form FP-RMS tumors in immunocompromised mice. Together these findings demonstrate that FP-RMS can originate from aberrant development of non-myogenic cells driven by P3F.
Collapse
Affiliation(s)
- Madeline B Searcy
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, 38105, USA
| | - Randolph K Larsen
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, 38105, USA
| | - Bradley T Stevens
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, 38105, USA
| | - Yang Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Catherine J Drummond
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Casey G Langdon
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Katherine E Gadek
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kyna Vuong
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Kristin B Reed
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Matthew R Garcia
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Darden W Kimbrough
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Rhodes College, Memphis, TN, 38112, USA
| | - Grace E Adkins
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN, 38105, USA
| | - Nadhir Djekidel
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shaina N Porter
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Patrick A Schreiner
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Brian J Abraham
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jerold E Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Mark E Hatley
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|