1
|
Sánchez-Vañó R, Balaguer J, Borrego-Dorado I, Esteban-Figueruelo A, Gámez C, Hladun R, López-Almaraz R, Llempén ML, Rodado S, Rubio-Aparicio PM. Recommendations for the use of nuclear medicine imaging in patients with neuroblastoma. Clin Transl Oncol 2025; 27:2401-2415. [PMID: 39508974 DOI: 10.1007/s12094-024-03755-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024]
Abstract
Neuroblastoma (NB) is the most common extracranial solid cancer in children. Despite intensive multimodality treatment, long-term survival of patients with high-risk NB, which comprises more than half of all cases, remains poor. Nuclear medicine is key in diagnosis, staging, response assessment and long-term follow-up of NB. The emergence of novel tracers and the increasing complexity of studies require updated guidelines for nuclear medicine imaging in NB. Standardising diagnostic techniques are essential for improving study comparability and ensuring test quality. This article aims to provide a comprehensive review of NB radionuclide diagnostic imaging, including its characteristics, accuracy, advantages, and limitations. It offers practical recommendations to multidisciplinary teams responsible for treating patients with NB. This review summarises the opinions of leading experts from the Neuroblastoma Spanish Group within the Spanish Society of Paediatric Haematology and Oncology (SEHOP) and the Spanish Society of Nuclear Medicine and Molecular Imaging (SEMNIM).
Collapse
Affiliation(s)
| | - Julia Balaguer
- Servicio de Hemato-Oncología Pediátrica, Hospital Universitario La Fe and Polytechnic Hospital, Valencia, Spain.
| | - Isabel Borrego-Dorado
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Cristina Gámez
- Servicio de Medicina Nuclear, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Raquel Hladun
- Servicio de Hemato-Oncología Pediátrica, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - Ricardo López-Almaraz
- Unidad de Hemato-Oncología Pediátrica, Hospital Universitario Cruces and Pediatric Oncology Group Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Mercedes L Llempén
- Servicio de Oncología Pediátrica, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Sonia Rodado
- Servicio de Medicina Nuclear, Hospital Universitario La Paz, Madrid, Spain
| | | |
Collapse
|
2
|
Kushner BH, Modak S, Mauguen A, Basu EM, Kramer K, Roberts SS, Cheung IY, Cheung NKV. A Phase II Trial of Naxitamab plus Stepped-up Dosing of GM-CSF for Patients with High-Risk Neuroblastoma in First Complete Remission. Clin Cancer Res 2025; 31:1877-1884. [PMID: 40067131 DOI: 10.1158/1078-0432.ccr-24-3427] [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: 10/16/2024] [Revised: 12/02/2024] [Accepted: 03/07/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE Naxitamab is a humanized form of the murine anti-GD2 mAb 3F8. In an international trial, naxitamab + GM-CSF was effective against chemoresistant high-risk neuroblastoma (HR-NB), leading to approval by the FDA. We now report results with patients in first complete remission (CR). PATIENTS AND METHODS The primary objective of this phase II protocol 16-1643 (Clinicaltrials.gov NCT03033303) was to assess event-free survival of patients with HR-NB in first CR treated with naxitamab + GM-CSF plus isotretinoin. HR-NB was defined as MYCN-amplified disease (any age) or metastatic disease at age >18 months. Cycles of immunotherapy were administered monthly up to five cycles and comprised (i) subcutaneously administered priming doses of GM-CSF 250 μg/m2/day on days -4 to -0 (Wednesday-Sunday), followed by a step-up to 500 μg/m2/day on days +1 to +5 (Monday-Friday) and (ii) naxitamab infused intravenously (30-90") on days +1, +3, and +5 (Monday-Wednesday-Friday, i.e., three doses/cycle). The dosage of naxitamab was 3 mg/kg/infusion (9 mg/kg/cycle, i.e., ∼270 mg/m2/cycle). The dosage of isotretinoin was 160 mg/m2/day started after cycle 2, ×14 days/course, and ×6 courses. RESULTS Fifty-nine patients with HR-NB (53 stage 4, 6 stage 3) were enrolled from February 2017 to July 2020. At 36 months, event-free/overall survival rates were 73%/93%, but 50 of 59 patients received after protocol treatment (vaccine and/or difluoromethylornithine). Six of 18 relapses were isolated in the central nervous system. Longer time from diagnosis to enrollment was a significantly adverse prognostic factor (P = 0.04). Twenty-one of 59 patients took no isotretinoin. Treatment was tolerable allowing outpatient administration. CONCLUSIONS Naxitamab + GM-CSF is a good option to consolidate first CR of patients with HR-NB, including those who did not undergo autologous stem-cell transplantation. Efforts to prevent central nervous system relapse are warranted.
Collapse
Affiliation(s)
- Brian H Kushner
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Audrey Mauguen
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ellen M Basu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kim Kramer
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stephen S Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene Y Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
3
|
Xie W, Zhang Y, Xu J, Sun F, Zhu J, Que Y, Huang J, Zhen Z, Lu S, Wang J, Zhang Y. Characteristics, treatments, and outcomes of adolescents and adults with neuroblastoma: a retrospective study in China. Ther Adv Med Oncol 2025; 17:17588359251337494. [PMID: 40351327 PMCID: PMC12064894 DOI: 10.1177/17588359251337494] [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: 10/27/2024] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
Background Neuroblastoma (NB) is rare in adolescents and adults, resulting in limited availability of data. Objectives We comprehensively investigated the characteristics, treatments, and outcomes of adolescent and adult patients with NB, aiming to provide a more in-depth insight into this disease. Design A retrospective, single-center study. Methods We retrieved and analyzed the medical data of patients with NB aged 10 years or older at diagnosis who were treated at Sun Yat-sen University Cancer Center between June 2005 and January 2024. Results Sixty-five patients (30 males and 35 females) were enrolled, with a median age of 20 years (interquartile range, 14-26 years), including 27 patients aged 10-18 years and 38 patients aged >18 years. Most patients were classified as M-stage disease (n = 40, 61.5%), high-risk (n = 42, 64.6%), and poorly differentiated NB (n = 27, 41.5%). Additionally, 3 (6.7%) patients had MYCN amplification, and 5 (25%) had ALK mutations. The genomic landscape revealed that mutations in the cell cycle and DNA repair pathways are related to chemotherapy sensitivity. After induction therapy, 34 (52.3%) patients achieved complete response (CR). The 5-year progression-free survival (PFS) and overall survival (OS) rates were 33.1% ± 6.9% and 55.1% ± 7.6%, respectively. Patients who achieved CR after induction therapy had superior PFS (p = 0.009), with 5-year PFS rates of 44.0% ± 10.6% compared to 18.5% ± 8.5% in non-CR patients. Conclusion Adolescent and adult patients with NB exhibit distinct characteristics, less chemotherapy sensitivity, and poorer outcomes compared to pediatric patients. Achieving CR after induction therapy is associated with better outcomes. Further investigation for new therapies is required.
Collapse
Affiliation(s)
- Weiji Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yu Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jiaqian Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Feifei Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jia Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yi Que
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Junting Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Zijun Zhen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Suying Lu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Yuexiu District, Guangzhou City, Guangdong 510060, P.R. China
| | - Juan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Yuexiu District, Guangzhou City, Guangdong 510060, P.R. China
| | - Yizhuo Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
- Department of Pediatric Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Yuexiu District, Guangzhou City, Guangdong 510060, P.R. China
| |
Collapse
|
4
|
Yu W, Biyik-Sit R, Uzun Y, Chen CH, Thadi A, Sussman JH, Pang M, Wu CY, Grossmann LD, Gao P, Wu DW, Yousey A, Zhang M, Turn CS, Zhang Z, Bandyopadhyay S, Huang J, Patel T, Chen C, Martinez D, Surrey LF, Hogarty MD, Bernt K, Zhang NR, Maris JM, Tan K. Longitudinal single-cell multiomic atlas of high-risk neuroblastoma reveals chemotherapy-induced tumor microenvironment rewiring. Nat Genet 2025; 57:1142-1154. [PMID: 40229600 DOI: 10.1038/s41588-025-02158-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025]
Abstract
High-risk neuroblastoma, a leading cause of pediatric cancer mortality, exhibits substantial intratumoral heterogeneity, contributing to therapeutic resistance. To understand tumor microenvironment evolution during therapy, we longitudinally profiled 22 patients with high-risk neuroblastoma before and after induction chemotherapy using single-nucleus RNA and ATAC sequencing and whole-genome sequencing. This revealed profound shifts in tumor and immune cell subpopulations after therapy and identified enhancer-driven transcriptional regulators of neuroblastoma neoplastic states. Poor outcome correlated with proliferative and metabolically active neoplastic states, whereas more differentiated neuronal-like states predicted better prognosis. Proportions of mesenchymal neoplastic cells increased after therapy and a high proportion correlated with a poorer chemotherapy response. Macrophages significantly expanded towards pro-angiogenic, immunosuppressive and metabolic phenotypes. We identified paracrine signaling networks and validated the HB-EGF-ERBB4 axis between macrophage and neoplastic subsets, which promoted tumor growth through the induction of ERK signaling. These findings collectively reveal intrinsic and extrinsic regulators of therapy response in high-risk neuroblastoma.
Collapse
Affiliation(s)
- Wenbao Yu
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rumeysa Biyik-Sit
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yasin Uzun
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Chia-Hui Chen
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anusha Thadi
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan H Sussman
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Minxing Pang
- Applied Mathematics and Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Chi-Yun Wu
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Liron D Grossmann
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Hemato-Oncology Division, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel HaShomer, Israel
- Cancer Research Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Peng Gao
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Genome Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - David W Wu
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aliza Yousey
- Center for Single Cell Biology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mei Zhang
- Center for Single Cell Biology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christina S Turn
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhan Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Shovik Bandyopadhyay
- Medical Scientist Training Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeffrey Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tasleema Patel
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Changya Chen
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Daniel Martinez
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lea F Surrey
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael D Hogarty
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kathrin Bernt
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nancy R Zhang
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - John M Maris
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kai Tan
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Center for Single Cell Biology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
5
|
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
|
6
|
Singh L, Iyer VK, Damle NA, Meena J, Seth R, Gupta A, Kakkar A, Rastogi K, Ganapathy C. Evaluating Bone Marrow Involvement in Pediatric Neuroblastoma: Traditional Methods and Emerging Technologies-A Systematic Review. Pediatr Dev Pathol 2025; 28:151-163. [PMID: 40159635 DOI: 10.1177/10935266251325632] [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] [Indexed: 04/02/2025]
Abstract
Neuroblastoma (NB) is the most common extracranial solid neoplasm affecting the pediatric population. It shows a high prevalence of bone marrow infiltration (BMI), which substantially impacts the disease's staging and prognostic assessment. Conventional methodologies, including bone marrow biopsy (BMB) and aspirate (BMA), have been extensively employed; nevertheless, the advent of novel technologies presents a promising avenue for diagnostic accuracy. This systematic review is designed to critically analyze and compare the established techniques (BMB and BMA) versus novel diagnostic approaches-such as immunocytology, RT-qPCR, and multiparametric flow cytometry (FCM), along with functional imaging like MIBG scintigraphy and FDG-PET/CT-in assessing BMI in pediatric NB. An exhaustive search was performed across the PubMed and Embase databases, identifying 2694 scholarly articles. Following a meticulous screening process and the application of inclusion criteria centered on diagnostic accuracy, sensitivity, and specificity about BMI, a total of 140 articles were selected for qualitative analysis. While BMB remains the gold standard for diagnosing and staging BMI in NB, recent advances in molecular techniques and functional imaging have shown superior sensitivity and specificity. Immunocytology and RT-qPCR can detect minimal residual disease (MRD) with higher sensitivity compared to traditional methods. Functional imaging modalities, particularly FDG-PET/CT and MIBG scintigraphy, have demonstrated improved accuracy in assessing bone marrow involvement with the added advantage of evaluating the entire bone marrow, overcoming the limitations of focal sampling in BMB. The integration of advanced molecular diagnostics and functional imaging with traditional biopsy methods enhances the accuracy of BMI in NB.
Collapse
Affiliation(s)
- Lavleen Singh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Venkateswaran K Iyer
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Nishikant A Damle
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Jagdish Meena
- Department of Pediatric Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Rachna Seth
- Department of Pediatric Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Aditya Gupta
- Department of Pediatric Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Aanchal Kakkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kanika Rastogi
- Department of Pathology, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, New Delhi, India
| | | |
Collapse
|
7
|
De Ioris MA, Villani MF, Fabozzi F, Del Bufalo F, Altini C, Cefalo MG, Cannata V, Del Baldo G, Pizzoferro M, Alessi I, Lanzaro F, Davide C, Tomà P, D'Andrea ML, Di Giannatale A, Serra A, Mastronuzzi A, Garganese MC, Locatelli F. 131I-mIBG therapy in relapsed/refractory neuroblastoma: an old bridge to the future. ESMO Open 2025; 10:104541. [PMID: 40187111 PMCID: PMC12002777 DOI: 10.1016/j.esmoop.2025.104541] [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: 09/26/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The prognosis of relapsed/refractory (R/R) neuroblastoma (NB) is still dismal. The role of iodine-131 meta-iodobenzylguanidine (131I-mIBG) treatment as a tool to reduce tumour burden before novel immunotherapies is not defined. PATIENTS AND METHODS Patients with R/R NB were included in a prospective observational study based on two infusions of 131I-mIBG plus melphalan (110 mg/m2), supported by autologous haematopoietic stem cell rescue. The activity of the first administration was 444 MBq (12 mCi/kg), while the second dose was modulated to reach a whole-body absorbed dose of 4 Gy. The International Neuroblastoma Response Criteria (INRC) were used for response. RESULTS Twenty-six patients with a median age of 5.9 years (range 2.5-17.2 years) were treated. Twenty-three patients presented a bone/bone marrow involvement, and 21 patients presented an uptake at primary site or at soft-tissue sites. The median International Society of Paediatric Oncology Europe Neuroblastoma Group (SIOPEN) skeletal score was 10 (range 1-70). The main recorded toxicities were haematological, with no toxic deaths and only one grade 4 mucositis. Hypothyroidism was reported in 6 patients of the 14 alive patients. The overall response rate was 48% [95% confidence interval (CI) 28% to 69%] with only one progression; after treatment the median SIOPEN skeletal score was 6 (range 0-70) with a median reduction of 35% (range 4.3%-100%). Overall, 52% (95% CI 32% to 73%) of patients achieved/maintained a SIOPEN skeletal score <7 and a soft-tissue lesion <5 cm was seen in 67% (95% CI 43% to 91%). After this treatment, 65% of patients underwent GD2-targeting chimeric antigen receptor (CAR)-T-cell therapy and 50%, high-dose chemotherapy with busulfan and melphalan. The 3-year overall survival was 55% (95% CI 33% to 73%) and event-free survival was 42% (95% CI 23% to 60%). CONCLUSION The 131I-mIBG therapy plus melphalan is confirmed to be effective to reduce/control tumour burden. Further studies are needed to clarify the role and timing of this treatment and to integrate its role in the strategy of CAR-T cells.
Collapse
Affiliation(s)
- M A De Ioris
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - M F Villani
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - F Fabozzi
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - F Del Bufalo
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - C Altini
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - M G Cefalo
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - V Cannata
- Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Radiology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - G Del Baldo
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - M Pizzoferro
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - I Alessi
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - F Lanzaro
- Department of Woman, Child and General and Specialized Surgery, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - C Davide
- Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - P Tomà
- Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - M L D'Andrea
- Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - A Di Giannatale
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - A Serra
- Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - A Mastronuzzi
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - M C Garganese
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - F Locatelli
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
8
|
Gawne PJ, Bryant HE, DuBois SG, George SL, Gray J, Knox L, Matchett KB, Peet C, Vallis KA, Wallace HJ, Wan S, Gaze MN. Theranostics for Neuroblastoma: Making Molecular Radiotherapy Work Better. J Nucl Med 2025; 66:490-496. [PMID: 39978816 PMCID: PMC11960609 DOI: 10.2967/jnumed.124.269121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Despite improvements in neuroblastoma treatment, survival figures lag behind those of many other childhood malignancies. New treatments, and better use of existing treatments, are essential to reduce mortality. Neuroblastoma expresses several molecular targets for radionuclide imaging and therapy, of which the most widely exploited is the norepinephrine transporter. [123I]metaiodobenzylguanidine (MIBG) imaging and [131I]MIBG treatment, which target this physiologic pathway, have been in clinical practice for 40 y. Although therapy outcomes have been favorable, [131I]MIBG use has not yet been optimized. Somatostatin receptors and the disialoganglioside are alternative targets, but their use remains experimental. The charity Children's Cancer Research Fund organized a workshop bringing together a broad range of scientists including radiochemists, radiobiologists, radiation physicists, clinical researchers including pediatric oncologists and nuclear medicine physicians, and patient advocates from the United Kingdom, United States, and continental Europe to share their experiences with molecular imaging and radiotherapy of neuroblastoma and discuss potential ways of improving treatment outcomes and access. These include development of alternative vectors targeting somatostatin receptors and disialoganglioside, isotopes such as α-particle and Auger electron emitters with different radiation characteristics, and combinations with external-beam radiotherapy, immunotherapy, and DNA damage repair inhibitors. Barriers to progress discussed included the unpredictable radioisotope supply, production of novel radiopharmaceuticals, lack of data regarding which are the best combination therapies, and insufficient clinical facilities. The aim was to stimulate the development and assessment of more effective treatments.
Collapse
Affiliation(s)
- Peter J Gawne
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary, University of London, London, United Kingdom
- UCL Cancer Institute, University College London, London, United Kingdom
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Helen E Bryant
- School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Sally L George
- Division of Clinical Studies, Institute of Cancer Research, London, United Kingdom
- Children and Young People's Unit, Royal Marsden Hospital, London, United Kingdom
| | - Juliet Gray
- Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
| | - Leona Knox
- Solving Kids' Cancer, London, United Kingdom
| | - Kyle B Matchett
- Personalised Medicine Centre and Clinical Translational Research and Innovation Centre, Altnagelvin Area Hospital, School of Medicine, Ulster University, Derry-Londonderry, United Kingdom
| | - Connie Peet
- Department of Radiotherapy, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | | | - Hugh J Wallace
- Paediatric Nuclear Medicine, Royal Hospital for Children, Glasgow, United Kingdom
- Department of Clinical Physics and Bioengineering, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- University of Glasgow, Glasgow, United Kingdom
| | - Simon Wan
- Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust/University College London, London, United Kingdom; and
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom @ucl.ac.uk
| |
Collapse
|
9
|
Yu X, Kang S, Ge J, Wang J. A clinical observational study of dinutuximab beta as first-line maintenance treatment for patients with high-risk neuroblastoma in China. BMC Pediatr 2025; 25:203. [PMID: 40091019 PMCID: PMC11912632 DOI: 10.1186/s12887-025-05568-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND High-risk neuroblastoma (HR-NB) is associated with high metastatic and relapse rates that require intensive multimodal treatment. We evaluated the efficacy and safety of dinutuximab beta as first-line maintenance immunotherapy in pediatric patients with HR-NB in real-world clinical settings in China. METHODS We retrospectively reviewed the clinical records of pediatric patients with newly diagnosed HR-NB in the hospital from October 2021 to November 2023. Patients treated with dinutuximab beta in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) and isotretinoin as the first-line maintenance therapy were included in this study. Among patients with residual disease after completing induction and consolidation treatment, those with partial response (PR) or very good partial response (VGPR) except for bone marrow (BM) residue were also administrated vincristine/irinotecan/temozolomide (VIT) chemotherapy. RESULTS Fifty-one patients with newly diagnosed HR-NB who achieved at least PR before immunotherapy were evaluated. At the end of immunotherapy, the objective response rate (ORR) in 33 patients with evidence of disease was 60.6% (95% confidence interval (CI), 42.1-77.1%) and the complete response rate (CRR; n = 18) was 54.5% (95% CI, 36.4-71.9%). The 2-year event-free survival (EFS) rate and overall survival (OS) rate were 80.1% (95% CI, 66.2-88.8%) and 97.6% (95% CI, 84.3-99.7%), respectively. The 2-year EFS rate was higher in patients with CR (94.4%; 95% CI, 66.6-99.2%) than in non-CR patients (72.6%; 95% CI, 53.9-84.7%). Dinutuximab beta was well tolerated in patients and had fewer side effects, which decreased over time. Co-treatment of dinutuximab beta with VIT chemotherapy did not require discontinuation in patients undergoing immunochemotherapy. CONCLUSION The study showed promising efficacy and safety of dinutuximab beta as the first-line maintenance immunotherapy for pediatric patients with HR-NB. Notably, the combination of dinutuximab beta with GM-CSF and VIT chemotherapy could be used for treating patients who did not achieve CR after previous multimodal therapy.
Collapse
Affiliation(s)
- Xuedi Yu
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Suyi Kang
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Junjie Ge
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Jingfu Wang
- Department of Pediatric Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250000, China.
| |
Collapse
|
10
|
Moraitis A, Prochnow A, Poeppel TD, Schmitz J, Laschinsky C, Herrmann K, Bockisch A, Fragoso Costa P, Kersting D, Jentzen W. Tumor Dose-Response Relationship of [ 131I]MIBG Therapy in Patients with Neural Crest Tumors by Means of [ 124I]MIBG PET. J Nucl Med 2025:jnumed.124.269377. [PMID: 40081949 DOI: 10.2967/jnumed.124.269377] [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: 12/16/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
[131I]Metaiodobenzylguanidine (MIBG) therapy in patients with neural crest tumors has demonstrated sustained control of catecholamine-associated hypertension and corresponding partial response. Details on how neural crest tumors respond to an absorbed dose delivered by [131I]MIBG-targeted therapies is insufficiently known. The primary aim of this retrospective study was to assess the tumor dose-response relationship by means of quantitative analysis of [124I]MIBG PET data. Methods: The tumor dose-response relationship was studied in patients with advanced malignant pheochromocytoma, neuroblastoma, or paraganglioma receiving [131I]MIBG treatment, as well as pretherapeutic and follow-up [124I]MIBG-based dosimetry. [124I]MIBG PET imaging was performed around 4, 24, 48, and 120 h after injection. Lesion uptake was projected to [131I]MIBG for every time point, and respective time-integrated activity coefficients (TIACs) for [131I]MIBG were calculated and used for tumor-absorbed dose estimation. Functional response was denoted for decrease of maximal lesion uptake or TIAC by at least 30% in the follow-up examination. In a consecutive analysis, the predictive value of a single tumor-uptake assessment from PET imaging at 24 h after administration was investigated with respect to receiving the derived target dose. Results: In total, 46 lesions from 9 patients were available for dose-response analysis. The mean ± SD tumor-absorbed dose coefficient was 13.4 ± 15.4 Gy/GBq (median, 7.2 Gy/GBq; range, 1.1-64.7 Gy/GBq). A high correlation (-0.60, P < 0.001) was found between uptake decrease and tumor dose. In addition, a very high correlation (0.91, P < 0.001) was found between uptake and TIAC decrease. The estimated targeted tumor dose was 200 Gy, that is, the dose at which the response rate exceeded the 90% threshold. A single 24-h uptake assessment showed predictive value with respect to receiving the target dose. Conclusion: This study demonstrated a clear correlation between tumor-absorbed dose and functional response in [131I]MIBG therapy and proposes a target dose for response at the tumor level.
Collapse
Affiliation(s)
- Alexandros Moraitis
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Andre Prochnow
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Thorsten Dirk Poeppel
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Jochen Schmitz
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Christina Laschinsky
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Andreas Bockisch
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Pedro Fragoso Costa
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - David Kersting
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Walter Jentzen
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
11
|
Wang X, Mao AW, Pan S, Wang D, He L, Vogel H, Mao JH, Weiss W, Li T, Chang H. Cellular morphometric biomarkers and large language model predict prognosis and treatment response in neuroblastoma patients: A retrospective and double-blind prospective single arm clinical study. Eur J Cancer 2025; 218:115273. [PMID: 39908653 DOI: 10.1016/j.ejca.2025.115273] [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: 05/25/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND The heterogeneity of Neuroblastoma (NB) leads to variation in response to treatment and outcomes. The aim of the current study is to discover AI-empowered cellular morphometric biomarkers (CMBs), to establish the corresponding CMB risk score (CMBRS), CMB risk group (CMBRG), large language model driven CMB risk score (CMB-LLM-RS), and large language model driven CMB risk group (CMB-LLM-RG), and to investigate and validate their prognostic and predictive power in NB. METHODS In this study, the retrospective cohort enrolled 84 primary NBs between 1/2020 and 12/2021, followed up through 11/22/2024; the prospective cohort enrolled 67 primary NBs between 1/2022 and 7/2023, followed up through 11/22/2024. RESULTS We identified 9 CMBs from a retrospective NB cohort, enabling the CMBRS, CMBRG, CMB-LLM-RS, and CMB-LLM-RG. Both CMBRG and CMB-LLM-RG are significantly associated with prognosis (p < 0.0001) and treatment response (p < 0.0001). Furthermore, we double-blindly validated the predictive power of CMBRG and CMB-LLM-RG in a prospective NB cohort, which confirms their potential value in real clinical settings. Importantly, CMBRG provides clinical value independent of the International Neuroblastoma Risk Group (INRG) classification system in both retrospective and prospective NB cohorts (p < 0.05); and the combination of CMBRG and INRG significantly increases prognostic and predictive performance for NB patients. CONCLUSIONS These findings suggest that CMBRG and CMB-LLM-RG have prognostic and predictive value for NB and warrants evaluation in larger multicenter cohorts.
Collapse
Affiliation(s)
- Xu Wang
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - April W Mao
- Department of Mathematics, University of California, Los Angeles, Los Angeles, United States
| | - Sirui Pan
- Department of Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Dawei Wang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Lili He
- Department of Pathology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hannes Vogel
- Department of Pathology, Stanford University Medical Center, Stanford, CA, United States
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - William Weiss
- Department of Neurology, Neurological Surgery, and Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Tao Li
- Department of Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States; Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
12
|
Quintarelli C, Del Bufalo F, De Ioris MA, Guercio M, Algeri M, Pagliara D, Silvestris DA, Di Nardo M, Sinibaldi M, Di Cecca S, Iaffaldano L, Manni S, Fustaino V, Garganese MC, Colafati GS, Bertaina V, Becilli M, Mastronuzzi A, Fabozzi F, Gunetti M, Iacovelli S, Bugianesi R, Macchia S, Li Pira G, Cefalo MG, Leone G, Del Baldo G, De Angelis B, Locatelli F. Donor-derived GD2-specific CAR T cells in relapsed or refractory neuroblastoma. Nat Med 2025; 31:849-860. [PMID: 39815015 DOI: 10.1038/s41591-024-03449-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025]
Abstract
Allogeneic chimeric antigen receptor (CAR) T cells targeting disialoganglioside-GD2 (ALLO_GD2-CART01) could be a therapeutic option for patients with relapsed or refractory, high-risk neuroblastoma (r/r HR-NB) whose tumors did not respond to autologous GD2-CART01 or who have profound lymphopenia. We present a case series of five children with HR-NB refractory to more than three different lines of therapy who received ALLO_GD2-CART01 in a hospital exemption setting. Four of them had previously received allogeneic hematopoietic stem cell transplantation. All patients experienced grade 2 or 3 cytokine release syndrome and one grade 2 neurotoxicity. Moderate acute graft-versus-host-disease occurred in four patients. ALLO_GD2-CART01 persisted for >6 weeks. Post-treatment, two complete responses were achieved and one maintained; in addition, one partial response and one stable disease were observed. Comparing the transcriptomic profiles obtained by RNA sequencing analyses of drug products with patient-matched, peripheral blood ALLO_GD2-CART01 collected at expansion, we found upregulation of genes associated with T cell activation and migration. In addition, after infusion, transcriptomic signaling analysis showed enrichment of genes involved in response to decreased oxygen levels, humoral immune response, cell polarization and immune-synapse formation. In comparison to autologous CAR T cells, ALLO_GD2-CAR T cells were characterized by pathways associated with T cell proliferation, immune-synapse formation and cell chemotaxis. The safety and efficacy of ALLO_GD2-CART01 in children with r/r HR-NB deserve further investigation in a prospective trial.
Collapse
Affiliation(s)
- Concetta Quintarelli
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesca Del Bufalo
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Antonietta De Ioris
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Marika Guercio
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Mattia Algeri
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daria Pagliara
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Domenico Alessandro Silvestris
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Matteo Di Nardo
- Pediatric Intensive Care Unit, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - Matilde Sinibaldi
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Stefano Di Cecca
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Laura Iaffaldano
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Simona Manni
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Valentina Fustaino
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Carmen Garganese
- Nuclear Medicine Unit/Imaging Department, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Valentina Bertaina
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Marco Becilli
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Angela Mastronuzzi
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Fabozzi
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Monica Gunetti
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Stefano Iacovelli
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Rossana Bugianesi
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Stefania Macchia
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Giuseppina Li Pira
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Giuseppina Cefalo
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Giovanna Leone
- Transfusion Unit, Department of Laboratories, IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - Giada Del Baldo
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Biagio De Angelis
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Cell and Gene Therapy, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Bambino Gesù Children's Hospital, Rome, Italy.
- Catholic University of the Sacred Heart, Department of Life Sciences and Public Health, Rome, Italy.
| |
Collapse
|
13
|
Fabozzi F, Villani MF, Del Bufalo F, Altini C, Cannatà V, Davide C, Pizzoferro M, Drago M, D'Antonio F, Triumbari EKA, Di Giannatale A, Vennarini S, Mastronuzzi A, De Ioris MA, Garganese MC. 131I-mIBG therapy in relapsed/refractory neuroblastoma: A weapon from the future past. Crit Rev Oncol Hematol 2025; 207:104606. [PMID: 39732302 DOI: 10.1016/j.critrevonc.2024.104606] [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: 08/06/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in children, with variable outcomes ranging from spontaneous remission to high-risk cases often leading to relapse or refractory disease. Approximately 50 % of patients with NB have high-risk features, often experiencing relapse or refractory disease despite intensive treatments and the prognosis remains poor, with long-term event-free survival (EFS) rates below 10 %,Radioactive iodine-labeled meta-iodobenzylguanidine (¹³¹I-mIBG) therapy, leveraging NB cells' radiosensitivity and expression of the norepinephrine transporter (NET), has shown promise in treating relapsed or refractory NB. Since 1985, ¹³¹I-mIBG has been studied to determine the maximum tolerated dose and side effects, with recent trials exploring its use in front-line treatment. Our systematic review, based on MEDLINE, EMBASE, and Cochrane CENTRAL databases up to December 2023, evaluates the effectiveness and toxicity of ¹³¹I-mIBG therapy in relapsed/refractory NB. It also discusses its potential role in conjunction with emerging therapies like CAR-T cells, haploidentical stem cell transplantation, and dinutuximab beta.
Collapse
Affiliation(s)
- Francesco Fabozzi
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Francesca Del Bufalo
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Claudio Altini
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Vittorio Cannatà
- Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Ciucci Davide
- Medical Physics Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Milena Pizzoferro
- Nuclear Medicine Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Margherita Drago
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Federica D'Antonio
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Angela Di Giannatale
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sabina Vennarini
- Pediatric Radiotherapy Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Angela Mastronuzzi
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Antonietta De Ioris
- Paediatric Haematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | | |
Collapse
|
14
|
Qian X, Zhang D, Li K, Chen W, Zhuang P, Wang H, Lei Z, Li Y, Eldridge J, Dong K, Zhai X. Adverse Reaction Reporting for Naxitamab in Chinese Expanded Access Treatment for Relapsed/Refractory High-Risk Neuroblastoma at the Children's Hospital of Fudan University. Drugs Real World Outcomes 2025; 12:115-123. [PMID: 39704915 PMCID: PMC11829859 DOI: 10.1007/s40801-024-00468-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND The humanized anti-disialoganglioside-2 monoclonal antibody naxitamab was approved in the USA in 2020 for the treatment of patients with relapsed/refractory high-risk neuroblastoma, limited to the bone or bone marrow, in combination with granulocyte-macrophage colony-stimulating factor. Treatment with naxitamab under expanded access was initiated by physicians from the Children's Hospital of Fudan University, Shanghai, China, in August 2021. OBJECTIVE We reviewed all suspected adverse reactions (ARs) reported to the Y-mAbs Argus Global Pharmacovigilance Safety Database for patients treated with naxitamab under expanded access in China from 1 August 2021 to 31 July 2022. METHODS We assessed patient demographics and the safety profile of naxitamab over multiple treatment cycles. RESULTS At the data cutoff, 41 patients with relapsed/refractory high-risk neuroblastoma had received a total of 150 treatment cycles (451 infusions) of naxitamab. The median number of cycles completed was three; 13 patients (32%) were receiving ongoing naxitamab treatment. The median patient age was 3 years (range 1-9 years) and 63% were female. Overall, ARs were reported in 89/150 cycles (59%); serious ARs were reported in 23/150 cycles (15%). The cumulative reporting rate (ARs/cycle) decreased after 3 versus 12 months of expanded access: all ARs (8.7-4.6), serious ARs (0.9-0.3), hypotension (1.4-1.0), flushing (0.7-0.5), cough (0.6-0.3), pain (0.5-0.2), and hypoxia (0.3-0.2). CONCLUSIONS During the first 12 months of expanded access treatment in China, 41 patients received naxitamab therapy with a cumulative 451 infusions administered. Over the course of this expanded access program, a reduction in the AR rate, including serious ARs, was observed as more patients were initiated and proceeded to later treatment cycles. While additional research is needed, the observed decrease in the AR rate may be attributed to clinicians' increased knowledge of AR management and hands-on experience with naxitamab-treated patients.
Collapse
Affiliation(s)
- Xiaowen Qian
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Dufei Zhang
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Kai Li
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Weiming Chen
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Peijun Zhuang
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Hongsheng Wang
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Zhixian Lei
- Hainan Branch of the Children's Hospital of Fudan University, Haikou, China
| | - Yan Li
- Hainan Branch of the Children's Hospital of Fudan University, Haikou, China
| | | | - Kuiran Dong
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China.
| | - Xiaowen Zhai
- Children's Hospital of Fudan University, 399 Wanyuan Road, Minhang District, Shanghai, China.
| |
Collapse
|
15
|
Pandit-Taskar N, Basu E, Balquin E, Mozley PD, Jacobson AF, Modak S. Safety observations in neuroblastoma patients undergoing 18 F- m FBG PET. Nucl Med Commun 2025; 46:245-247. [PMID: 39641193 DOI: 10.1097/mnm.0000000000001938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Limited safety data have been published on fluorine-18 ( 18 F) meta-fluorobenzylguanidine ( m FBG), a new PET radiopharmaceutical for imaging neural crest and neuroendocrine tumors. As part of a prospective clinical trial, safety data in patients with neuroblastoma were collected and analyzed. METHODS Between April 2015 and January 2022, 27 patients with neuroblastoma underwent 18 F- m FBG PET imaging as part of an ongoing single-center phase 1/2 trial (NCT02348749). Pre- and postinjection safety assessments were performed, including vital sign measurement and observation for occurrence of adverse events (AEs). RESULTS m FBG administration resulted in no significant changes in measured vital signs. Two subjects had transient, grade 1 facial flushing shortly after the administration, which resolved within a few minutes. Neither subject had a clinically significant change in pulse or blood pressure on postadministration measurements. CONCLUSION In this investigation of the potential clinical utility of m FBG PET imaging, no significant adverse safety signals were noted. Two mild, self-limited AEs were observed, without associated changes in vital signs. No grade 2 or higher AEs were noted. The findings are consistent with a favorable safety profile for m FBG in the target population of patients with neuroblastoma.
Collapse
Affiliation(s)
| | - Ellen Basu
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York City, New York
| | | | - P David Mozley
- Innervate Radiopharmaceuticals LLC, Englewood, New Jersey, USA
| | | | - Shakeel Modak
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York City, New York
| |
Collapse
|
16
|
de Las Heras BM, Rubio-Aparicio PM, Rubio-San-Simón A, Moreno L, Mazorra P, Almaraz RL, López ML, Guill JB, Segura V, Bermúdez M, Jiménez I, Ramal D, Cañete A. Management and outcome of children with high-risk neuroblastoma: insights from the Spanish Society of Pediatric Hematology and Oncology (SEHOP) neuroblastoma group on refractory and relapse/progressive disease. Clin Transl Oncol 2025:10.1007/s12094-025-03853-w. [PMID: 39998749 DOI: 10.1007/s12094-025-03853-w] [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: 09/05/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Outcome for children with refractory and relapse/progressive high-risk neuroblastoma (HR-NB) remains poor, without an internationally agreed standard second-line approach. Heterogeneity in patients' disease and treatment strategies challenges clinical management. The survival rate for patients with resistant disease does not exceed 20% at 5 years. The study's aim was to analyze refractory and progressive HR-NB patients in a real-world setting to evaluate current clinical practices and optimize future approaches. METHODS Data from patients diagnosed with refractory and relapse/progressive (R/R-P) HR-NB between January 2019 and December 2021 at six of the major Spanish neuroblastoma treating hospitals were collected and analyzed. RESULTS A total of 67 episodes of R/R-P HR-NB were included. Treatments applied included chemotherapy (97%), immunotherapy (48%), consolidation (21%), local treatment (surgery and/or radiotherapy) (45%) and maintenance (16%), and were administered within a clinical trial (CT) in 34% of the episodes. Biopsy was performed in 37% of the tumors and 30% were profiled. Event-free survival (EFS) in our cohort was 20.9% and overall survival (OS) 32%. Significant survival advantage (in both OS and EFS) was observed in refractory episodes compared to relapse/progressive, in first events compared to successive, and when response or disease stabilization was achieved. MYCN status, presence of lymph node metastases, use of irinotecan or topotecan, and radiotherapy were also univariate predictors of OS. CONCLUSIONS Treatment of refractory and relapse/progressive HR-NB is highly heterogeneous. We confirm a poor outcome, although certain epidemiological and treatment-related factors have prognostic value. Molecular profiling and inclusion in CTs should be improved.
Collapse
Affiliation(s)
- Blanca Martínez de Las Heras
- Pediatric Hemato-Oncology Department, Hospital Universitario y Politécnico La Fe, European Reference Network PAEDCAN member, Valencia, Spain.
- Clinical and Translational Oncology Research Group, Investigation Institute La Fe, Valencia, Spain.
| | - Pedro M Rubio-Aparicio
- Pediatric Hemato-Oncology Department, Hospital Universitario La Paz, European Reference Network PAEDCAN member, Madrid, Spain
| | - Alba Rubio-San-Simón
- Pediatric Hemato-Oncology Department, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Lucas Moreno
- Pediatric Department, Hospital Universitario Vall d´Hebron, European Reference Network PAEDCAN member, Barcelona, Spain
| | - Paula Mazorra
- Pediatric Department, Hospital Universitario Vall d´Hebron, European Reference Network PAEDCAN member, Barcelona, Spain
| | - Ricardo López Almaraz
- Pediatric Onco-Hematology Unit, Hospital Cruces, and Pediatric Oncology Group Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Mercedes Llempén López
- Pediatric Oncology Department, Hospital Universitario Virgen del Rocío, European Reference Network PAEDCAN member, Sevilla, Spain
| | - Julia Balaguer Guill
- Pediatric Hemato-Oncology Department, Hospital Universitario y Politécnico La Fe, European Reference Network PAEDCAN member, Valencia, Spain
- Clinical and Translational Oncology Research Group, Investigation Institute La Fe, Valencia, Spain
| | - Vanessa Segura
- Clinical and Translational Oncology Research Group, Investigation Institute La Fe, Valencia, Spain
| | - Mar Bermúdez
- Pediatric Hematology-Oncology Department, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Irene Jiménez
- Pediatric Hematology-Oncology Department, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Désirée Ramal
- Clinical and Translational Oncology Research Group, Investigation Institute La Fe, Valencia, Spain
| | - Adela Cañete
- Pediatric Hemato-Oncology Department, Hospital Universitario y Politécnico La Fe, European Reference Network PAEDCAN member, Valencia, Spain
- Clinical and Translational Oncology Research Group, Investigation Institute La Fe, Valencia, Spain
| |
Collapse
|
17
|
DuBois SG, Ogawa C, Moreno L, Mossé YP, Fischer M, Ryan AL, Vo KT, De Wilde B, Rubio-San-Simon A, Macy ME, Howell L, Shusterman S, Corradini N, Luksch R, Aerts I, Foster JH, Weiss BD, Karthik CP, Yuen E, Avsar E, Park JR, Marachelian A. A phase 1 dose-escalation study of LY3295668 erbumine as monotherapy and in combination with topotecan and cyclophosphamide in children with relapsed/refractory neuroblastoma. Cancer 2025; 131:e35751. [PMID: 39932800 DOI: 10.1002/cncr.35751] [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: 10/03/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND This study evaluated the safety, pharmacokinetics, and antitumor activity of LY3295668 erbumine as monotherapy and combination therapy in children with relapsed/refractory neuroblastoma. METHODS Patients aged 2-21 years who had relapsed/refractory neuroblastoma were enrolled. LY3295668 erbumine was evaluated at two dose levels (12 and 15 mg/m2) and administered orally twice daily continuously as monotherapy and in combination with intravenous topotecan and cyclophosphamide in 28-day cycles. RESULTS Twenty-five patients were treated. No dose-limiting toxicity occurred in monotherapy; one patient had dose-limiting toxicities in the combination therapy cohort (grade 3 mucositis and grade 4 neutropenia). The recommended phase 2 dose for both monotherapy and combination therapy was 15 mg/m2. Twenty-two patients (88%) had one or more treatment-related adverse event(s) (TRAEs), and 18 (72%) experienced grade ≥3 TRAEs. Myelosuppression was the most common high-grade TRAE observed in the combination therapy cohort. At both dose levels, steady-state plasma concentrations exceeded xenograft 90% inhibitory concentration levels. In the monotherapy cohort, one patient had a minor response, and one patient had stable disease, both continuing for >12 months. In the combination therapy cohort, two patients had a partial response, two had a minor response, and six had stable disease. Overall, the response rate, according to New Approaches to Neuroblastoma Therapy version 2.0 criteria, was 8%, and the disease control rate was 52%. CONCLUSIONS LY3295668 erbumine had a manageable safety profile as monotherapy and in combination therapy. Although proof-of-concept clinical responses were observed, future studies with biomarker-selected populations and/or novel combinations may yield higher response rates with Aurora kinase A inhibition.
Collapse
Affiliation(s)
- Steven G DuBois
- Department of Pediatric Hematology/Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Chitose Ogawa
- Department of Pediatric Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Lucas Moreno
- Pediatric Oncology and Hematology, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona/Vall d'Hebron University Hospital, Barcelona, Spain
| | - Yaël P Mossé
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, Children's Hospital and Center for Molecular Medicine Cologne, Medical Faculty, University of Cologne, Cologne, Germany
| | - Anne L Ryan
- Department of Hematology and Oncology, Perth Children's Hospital, Nedlands, West Australia, Australia
| | - Kieuhoa T Vo
- Department of Pediatrics, University of California, San Francisco Benioff Children's Hospital and San Francisco School of Medicine, San Francisco, California, USA
| | - Bram De Wilde
- Pediatric Hematology and Oncology, Ghent University Hospital, Ghent, Belgium
| | - Alba Rubio-San-Simon
- Pediatric Hematology-Oncology Department, Children's University Hospital Niño Jesús, Madrid, Spain
| | - Margaret E Macy
- Pediatric Hematology, Oncology, and Bone Marrow Transplant, Children's Hospital Colorado/University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lisa Howell
- Alder Hey Children's National Health Service Foundation Trust, Liverpool, UK
| | - Suzanne Shusterman
- Department of Pediatric Hematology/Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Nadège Corradini
- Pediatric Hematology and Oncology Institute, Léon Bérard Center, Lyon, France
| | - Roberto Luksch
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Isabelle Aerts
- SIREDO Oncology Center (Care, Innovation, and Research for Children and Adolescents and Young Adults with Cancer), Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Jennifer H Foster
- Department of Pediatric Oncology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Brian D Weiss
- Division of Hematology-Oncology/Stem Cell Transplant, Department of Pediatrics, Indiana University School of Medicine, Riley Children's Health, Indianapolis, Indiana, USA
| | | | | | | | - Julie R Park
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Araz Marachelian
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, Children's Hospital Los Angeles/Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
18
|
Mora J, Chan GCF, Morgenstern DA, Amoroso L, Nysom K, Faber J, Wingerter A, Bear MK, Rubio-San-Simon A, de Las Heras BM, Tornøe K, Düring M, Kushner BH. The anti-GD2 monoclonal antibody naxitamab plus GM-CSF for relapsed or refractory high-risk neuroblastoma: a phase 2 clinical trial. Nat Commun 2025; 16:1636. [PMID: 39952926 PMCID: PMC11828896 DOI: 10.1038/s41467-025-56619-x] [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: 01/19/2024] [Accepted: 01/20/2025] [Indexed: 02/17/2025] Open
Abstract
In this single-arm, non-randomized, phase 2 trial (NCT03363373), 74 patients with relapsed/refractory high-risk neuroblastoma and residual disease in bone/bone marrow (BM) received naxitamab on Days 1, 3, and 5 (3 mg/kg/day) with granulocyte-macrophage colony-stimulating factor (Days -4 to 5) every 4 weeks, until complete response (CR) or partial response (PR) followed by 5 additional cycles every 4 weeks. Primary endpoint in the prespecified interim analysis was overall response (2017 International Neuroblastoma Response Criteria). Among 26 responders (CR + PR) in the efficacy population (N = 52), 58% had refractory disease, and 42% had relapsed disease. Overall response rate (ORR) was 50% (95% CI: 36-64%), and CR and PR were observed in 38% and 12%, respectively. With the 95% CI lower limit for ORR exceeding 20%, the primary endpoint of overall response was met. Patients with evaluable bone disease had a 58% (29/50) bone compartment response (CR, 40%; PR, 18%). BM compartment response was 74% (17/23; CR, 74%). One-year overall survival and progression-free survival (secondary endpoints) were 93% (95% CI: 80-98%) and 35% (95% CI: 16-54%), respectively. Naxitamab-related Grade 3 adverse events included hypotension (58%) and pain (54%). Overall, naxitamab demonstrated clinically meaningful efficacy with manageable safety in patients with residual neuroblastoma in bone/BM.
Collapse
Affiliation(s)
- Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Barcelona, Spain.
| | - Godfrey C F Chan
- Queen Mary Hospital & Hong Kong Children's Hospital, Pok Fu Lam, Hong Kong
- The University of Hong Kong, Pok Fu Lam, Hong Kong
| | | | - Loredana Amoroso
- IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Maternal Infantile and Urological Sciences, Pediatric Onco-Hematology Unit, Policlinico Umberto I, Sapienza, University of Rome, Rome, Italy
| | - Karsten Nysom
- Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology/Hemostaseology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Arthur Wingerter
- Department of Pediatric Hematology/Oncology/Hemostaseology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
19
|
Seo ES, Jeong IH, Ju HY, Hyun JK, Lee JW, Yoo KH, Heo WY, Sung KW, Cho HW, Kang ES. Predicted indirectly recognizable HLA epitopes scores and clinical outcomes after haploidentical stem cell transplantation in pediatric patients with relapsed neuroblastoma. Front Immunol 2025; 16:1517387. [PMID: 39967668 PMCID: PMC11833256 DOI: 10.3389/fimmu.2025.1517387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction The Predicted Indirectly ReCognizable HLA Epitopes (PIRCHE) model is a recently developed algorithm that predicts indirect T-cell recognition by calculating the number of such epitopes in donor-recipient pairs. Methods In this study, the clinical significance of PIRCHE was evaluated in pediatric patients with relapsed/progressed neuroblastoma undergoing haploidentical stem cell transplantation (haplo-SCT). Results A higher PIRCHE-I score was associated with faster platelet recovery (P = 0.007) and lower incidence of hemorrhagic cystitis (13% vs. 41%, P = 0.028) and invasive fungal infections (0% vs. 18%, P = 0.045). Additionally, a higher PIRCHE-I score was significantly associated with better overall survival (OS) (HR 0.57, 95% CI 0.34-0.97, P = 0.038). A higher PIRCHE-II score was associated with better OS (HR 0.57, 95% CI 0.34-0.94, P = 0.028) and reduced progression (HR 0.48, 95% CI 0.30-0.77, P = 0.002). When combined, the PIRCHE-I and PIRCHE-II scores demonstrated an even stronger association with improved OS (HR 0.35, 95% CI 0.15-0.82, P = 0.016). Multivariable analysis confirmed that a higher combined PIRCHE-I and PIRCHE-II score was independently associated with improved OS (combined PIRCHE score HR 0.22, 95% CI 0.06-0.79, P = 0.021), and a higher PIRCHE-II score was significantly associated with reduced progression (HR 0.42, 95% CI 0.25-0.70, P < 0.001). Conclusion In conclusion, higher PIRCHE-I and PIRCHE-II scores are linked to better survival outcomes and reduced complications in pediatric haplo-SCT neuroblastoma patients. Incorporating PIRCHE scores into donor selection is expected to optimize transplant outcomes.
Collapse
Affiliation(s)
- Eun Seop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - In Hwa Jeong
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Laboratory Medicine, Dong-A University Hospital, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ju Kyung Hyun
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Young Heo
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Suk Kang
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Hollander JF, Szymansky A, Wünschel J, Astrahantseff K, Rosswog C, Thorwarth A, Thole-Kliesch TM, Chamorro González R, Hundsdörfer P, Hauptmann K, Schmelz K, Gürgen D, Rogasch JM, Henssen AG, Fischer M, Schulte JH, Eckert C, Eggert A, Lodrini M, Deubzer HE. Serially Quantifying TERT Rearrangement Breakpoints in ctDNA Enables Minimal Residual Disease Monitoring in Patients with Neuroblastoma. CANCER RESEARCH COMMUNICATIONS 2025; 5:167-177. [PMID: 39760332 PMCID: PMC11774142 DOI: 10.1158/2767-9764.crc-24-0569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
SIGNIFICANCE Real-time molecular monitoring of TERT-rearranged high-risk neuroblastoma is an unmet clinical need. We tested liquid biopsy-based assays for patient-individualized TERT breakpoint sequences to monitor disease in pediatric patients. Our digital PCR approach provides high resolution of spatial and temporal disease quantification in individual patients and is applicable for clinical routine.
Collapse
Affiliation(s)
- Jan F. Hollander
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Annabell Szymansky
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Jasmin Wünschel
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Carolina Rosswog
- Department of Experimental Pediatric Oncology, University Children’s Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Anne Thorwarth
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Theresa M. Thole-Kliesch
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Rocío Chamorro González
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of Charité, Max-Delbrück-Center of Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Max-Delbrück Center of Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Patrick Hundsdörfer
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatrics, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Kathrin Hauptmann
- Institute of Pathology, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karin Schmelz
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Charité - 3R, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Dennis Gürgen
- Experimental Pharmacology and Oncology Berlin-Buch GmbH (EPO), Berlin, Germany
| | - Julian M.M. Rogasch
- Department of Nuclear Medicine, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Berlin, Germany
| | - Anton G. Henssen
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of Charité, Max-Delbrück-Center of Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Max-Delbrück Center of Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Berlin, Germany
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University Children’s Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Johannes H. Schulte
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology and Hematology, University Hospital Tübingen, Tübingen, Germany
| | - Cornelia Eckert
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marco Lodrini
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Hedwig E. Deubzer
- Department of Pediatric Oncology and Hematology, Campus Virchow Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of Charité, Max-Delbrück-Center of Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Max-Delbrück Center of Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
21
|
Matser YA, Samim A, Fiocco M, van de Mheen M, van der Ham M, de Sain-van der Velden MG, Verhoeven-Duif NM, van Grotel M, Kraal KC, Dierselhuis MP, van Eijkelenburg NK, Langenberg KP, van Noesel MM, van Kuilenburg AB, Tytgat GA. Urinary Catecholamines Predict Relapse During Complete Remission in High-Risk Neuroblastoma. JCO Precis Oncol 2025; 9:e2400491. [PMID: 39983076 PMCID: PMC11867808 DOI: 10.1200/po-24-00491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/23/2024] [Accepted: 01/06/2025] [Indexed: 02/23/2025] Open
Abstract
PURPOSE Urinary catecholamine metabolites are well-known biomarkers for the diagnosis (Dx) of neuroblastoma, but their clinical significance in determining therapy response during treatment is not well established. Therefore, catecholamines are not included in criteria for assessing response and complete remission (CR). This study investigated the use of urinary catecholamines in response monitoring and predicting survival outcomes. METHODS From 2005 to 2021, a panel of eight urinary catecholamines were measured in patients with high-risk neuroblastoma at Dx and at standard evaluation moments during treatment. At the same time points, response and CR were assessed according to the revised International Neuroblastoma Response Criteria. RESULTS The total cohort consists of 153 high-risk patients, and at least one of the eight metabolites was elevated (ie, catecholamine status positive) in 141 of 146 (97%), 104 of 128 (81%), and 39 of 69 (57%) patients at Dx, postinduction, and at CR, respectively. Primary tumor resection significantly reduced catecholamine levels (P < .01). A positive catecholamine status at Dx, during treatment, and at the end of treatment was not significantly associated with event-free survival (EFS) or overall survival (OS). However, in patients who achieved CR, those with a positive catecholamine status had poor EFS (38% v 80%, respectively; P < .01) and OS (52% v 86%, respectively; P = .01) compared with those with a negative catecholamine status. Notably, 3-methoxytyramine levels at CR seem to be a prognostic marker for poor OS (hazard ratio, 7.5 [95% CI, 2.0 to 28.6]). CONCLUSION Catecholamine measurements contribute to the assessment of CR and identifies patients with high-risk neuroblastoma with an increased risk of relapse and death.
Collapse
Affiliation(s)
| | - Atia Samim
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center, Utrecht, the Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Biomedical Data Science, Section Medical Statistics, Leiden University Medical Centre, Leiden, the Netherlands
- Mathematical Institute, Leiden University, Leiden, the Netherlands
| | | | - Maria van der Ham
- Department of Genetics, Section Metabolic Diagnostics, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | | | - Nanda M. Verhoeven-Duif
- Department of Genetics, Section Metabolic Diagnostics, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | | | | | | | | | | | - Max M. van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Division of Imaging and Oncology, University Medical Center, Utrecht, the Netherlands
| | - André B.P. van Kuilenburg
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Free University Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands
| | - Godelieve A.M. Tytgat
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Department of Genetics, Utrecht University Medical Center, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
22
|
van Zogchel LMJ, Decarolis B, van Wezel EM, Zappeij-Kannegieter L, Gelineau NU, Schumacher-Kuckelkorn R, Simon T, Berthold F, van Noesel MM, Fiocco M, van der Schoot CE, Hero B, Stutterheim J, Tytgat GAM. Sensitive liquid biopsy monitoring correlates with outcome in the prospective international GPOH-DCOG high-risk neuroblastoma RT-qPCR validation study. J Exp Clin Cancer Res 2024; 43:331. [PMID: 39722049 DOI: 10.1186/s13046-024-03261-y] [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: 11/06/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Liquid biopsies offer less burdensome sensitive disease monitoring. Bone marrow (BM) metastases, common in various cancers including neuroblastoma, is associated with poor outcomes. In pediatric high-risk neuroblastoma most patients initially respond to treatment, but in the majority the disease recurs with only 40% long-term survivors, stressing the need for more sensitive detection of disseminated disease during therapy. METHODS To validate sensitive neuroblastoma mRNA RT-qPCR BM testing, we prospectively assessed serial BM samples from 345 international high-risk neuroblastoma patients, treated in trials NB2004 (GPOH) or NBL2009 (DCOG), using PHOX2B, TH, DDC, CHRNA3, and GAP43 RT-qPCR mRNA markers and BM GD2-immunocytology. Association between BM-infiltration levels and event-free survival (EFS) and overall survival (OS) was estimated by using Cox regression models and Kaplan-Meier's methodology. RESULTS BM infiltration >10% by RT-qPCR at diagnosis was prognostic for survival (adjusted hazard ratio (HR) 1.82 [95%CI 1.25-2.63] and 2.04 [1.33-3.14] for EFS and OS, respectively). Any post-induction RT-qPCR positivity correlated with poor EFS and OS, with a HR of 2.10 [1.27-3.49] and 1.76 [1.01-3.08] and 5-years EFS of 26.6% [standard error 5.2%] versus 60.4% [6.7] and OS of 43.8% [5.9] versus 65.7% [6.6] for RT-qPCR-positive patients versus RT-qPCR-negative patients. In contrast, post-induction immunocytology positivity was not associated with EFS or OS (HR 1.22 [0.68-2.19] and 1.26 [0.54-2.42]). CONCLUSION This study validates the association of not clearing of BM metastases by sensitive RT-qPCR detection with very poor outcome. We therefore propose implementation of RT-qPCR for minimal residual disease testing in neuroblastoma to guide therapy.
Collapse
Affiliation(s)
- Lieke M J van Zogchel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Boris Decarolis
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Esther M van Wezel
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Lily Zappeij-Kannegieter
- Department of Immunocytology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Nina U Gelineau
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Roswitha Schumacher-Kuckelkorn
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Thorsten Simon
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Frank Berthold
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | - Max M van Noesel
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Division Imaging & Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Department of Biomedical data Science, Section Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, University Children's Hospital of Cologne, and Medical Faculty, University of Cologne, Köln, Germany
| | | | - Godelieve A M Tytgat
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory of the Amsterdam UMC, Amsterdam, The Netherlands.
- Department of Genetics, Utrecht University Medical Center, Utrecht, the Netherlands.
| |
Collapse
|
23
|
Lilley MM, Salek M, Holland A, Inaba H, Federico SM. Treatment of a pediatric patient with concurrent neuroblastoma and acute lymphoblastic leukemia. Pediatr Blood Cancer 2024; 71:e31313. [PMID: 39289847 DOI: 10.1002/pbc.31313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024]
Affiliation(s)
- Megan M Lilley
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Marta Salek
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Ashley Holland
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Hiroto Inaba
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sara M Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
24
|
Sokol E, LaBarre B, Pinto N, Kreissman S, Granger MM, Park JR, Bagatell R, Naranjo A, DuBois SG. Response to induction chemotherapy modifies the effect of conventional prognostic factors in high-risk neuroblastoma: A report from the Children's Oncology Group. EJC PAEDIATRIC ONCOLOGY 2024; 4:100193. [PMID: 39822770 PMCID: PMC11737523 DOI: 10.1016/j.ejcped.2024.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Background Response to induction chemotherapy has been shown to predict outcome in patients with high-risk neuroblastoma (HR-NB), with those achieving a complete response (CR) having superior outcomes. Methods We evaluated whether conventional prognostic factors remain prognostic in subsets of patients defined by response to induction. 1244 Patients from four COG high-risk trials were included. End-induction response was coded as CR, partial response (PR) or better, less than PR without progressive disease (PD), and PD. Cox regression models were performed to calculate event-free and overall survival (EFS, OS) hazard ratios, including interaction terms between induction response and prognostic factors including sex, age, stage, primary tumor location, LDH, ferritin, ploidy, MYCN status, ALK status, histology, MKI, grade, and study era. Results Among patients who achieved a CR after induction, INSS stage 4 disease and trial era were the only factors that remained significantly associated with inferior OS. For those who achieved less than a PR, adrenal primary site, MYCN amplification, and 1p LOH were associated with inferior outcomes. Multivariable models showed that end-induction response remained prognostic of EFS and OS even after controlling for other factors. Multiple significant statistical interactions were observed between end-induction response and other prognostic factors. Conclusion The impact of conventional prognostic factors is not static in patients with HR-NB. Instead, response to induction chemotherapy modifies the effect of conventional prognostic factors. These data can help to further refine prognosis for patients with variable responses to induction and help to identify candidates who might benefit from treatment other than standard post-induction therapy.
Collapse
Affiliation(s)
- Elizabeth Sokol
- Division of Pediatric Hematology, Oncology, Neuro-oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Brian LaBarre
- Department of Biostatistics, Children’s Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL, USA
| | - Navin Pinto
- Department of Pediatrics, Children’s Hospital of Colorado, University of Colorado School of Medicine, Aurora, CO, USA
| | - Susan Kreissman
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Duke University Medical Center, Durham, NC, USA
| | - M. Meaghan Granger
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Julie R. Park
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rochelle Bagatell
- Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Arlene Naranjo
- Department of Biostatistics, Children’s Oncology Group Statistics and Data Center, University of Florida, Gainesville, FL, USA
| | - Steven G. DuBois
- Dana-Farber / Boston Children’s Cancer and Blood Disorders Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
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
|
26
|
Wang H, Cai J. Quantitative MRI in Childhood Neuroblastoma: Beyond the Assessment of Image-defined Risk Factors. Radiol Imaging Cancer 2024; 6:e240089. [PMID: 39485111 PMCID: PMC11615636 DOI: 10.1148/rycan.240089] [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: 03/28/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024]
Abstract
Neuroblastoma commonly occurs in children. MRI is a radiation-free imaging modality and has played an important role in identifying image-defined risk factors of neuroblastoma, providing necessary guidance for surgical resection and treatment response evaluation. However, image-defined risk factors are limited to providing structural information about neuroblastoma. With the evolution of MRI technologies, quantitative MRI can not only help assess image-defined risk factors but can also quantitatively reflect the biologic features of neuroblastoma in a noninvasive manner. Therefore, compared with anatomic imaging, these emerging quantitative MRI technologies are expected to provide more imaging biomarkers for the management of neuroblastoma. This review article discusses the current applications of quantitative MRI technologies in evaluating childhood neuroblastoma. Keywords: Pediatrics, MR-Functional Imaging, Children, MRI, Neuroblastoma, Quantitative Imaging © RSNA, 2024.
Collapse
Affiliation(s)
- Haoru Wang
- From the Department of Radiology, Children’s Hospital of
Chongqing 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 Child Neurodevelopment and Cognitive
Disorders, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014,
China
| | - Jinhua Cai
- From the Department of Radiology, Children’s Hospital of
Chongqing 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 Child Neurodevelopment and Cognitive
Disorders, No. 136 Zhongshan Road 2, Yuzhong District, Chongqing 400014,
China
| |
Collapse
|
27
|
Lu X, Li C, Wang S, Yin Y, Fu H, Wang H, Cheng W, Chen S. The prognostic role of 18F-FDG PET/CT-based response evaluation in children with stage 4 neuroblastoma. Eur Radiol 2024; 34:7125-7135. [PMID: 38758254 DOI: 10.1007/s00330-024-10781-w] [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: 11/16/2023] [Revised: 03/11/2024] [Accepted: 04/04/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVES To evaluate the association between metabolic response on 18F-FDG PET/CT and long-term survival in children with neuroblastoma (NB). METHODS A total of 39 consecutive children with newly diagnosed stage 4 NB undergoing both 18F-FDG PET/CT imaging at baseline and after chemotherapy were retrospectively analyzed. The associations between metabolic parameters, including SUVmax of the lesion with the most intense 18F-FDG uptake at baseline (SUVb), after chemotherapy (SUVe), and the percentage change between SUVb and SUVe, and long-term survival were evaluated. RESULTS With a median follow-up of 56 months, 22 patients who had achieved complete resolution on PET (no residual 18F-FDG uptake higher than the surrounding backgrounds) after chemotherapy had superior 5-year overall survival (OS) (73.6% vs. 39.0%, p = 0.044). SUVb > 6.9 indicated significantly poorer 5-year event-free survival (EFS) (12.5% vs. 59.3%, p = 0.005), as did SUVe > 1.2 (18.8% vs. 41.7%, p = 0.041). Children with SUVe > 1.2 had shorter 5-year OS (33.9% vs. 75.0%, p = 0.018). Multivariate analysis identified SUVe > 1.2 as an independent predictor for both EFS [hazard ratio (HR), 3.479, 95% CI, 1.381-8.761, p = 0.008] and OS (HR, 6.948, 95% CI, 1.663-29.025, p = 0.008), while SUVb > 6.9 was a predictor for EFS (HR, 2.889, 95% CI, 1.064-7.842, p = 0.037). Among 11 children with both SUVb > 6.9 and SUVe > 1.2, all experienced disease progression or relapse within 2 years since diagnosis. CONCLUSION 18F-FDG PET/CT could be of useful to evaluate treatment response in children with stage 4 NB. CLINICAL RELEVANCE STATEMENT 18F-FDG PET/CT after chemotherapy exhibits prognostic significance in neuroblastoma and holds potential as an alternative imaging modality for response evaluation, especially in cases with metaiodobenzylguanidine-nonavid or persistent avid disease. KEY POINTS The prognostic value of chemotherapy response on 18F-FDG PET/CT in advanced neuroblastoma is unknown. Higher 18F-FDG uptake after chemotherapy was associated with worse long-term event-free survival and overall survival. 18F-FDG PET/CT after chemotherapy holds prognostic significance in children with stage 4 neuroblastoma.
Collapse
Affiliation(s)
- Xueyuan Lu
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Li
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoyan Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongliang Fu
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Cheng
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Suyun Chen
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
28
|
Castelli S, Thorwarth A, van Schewick C, Wendt A, Astrahantseff K, Szymansky A, Lodrini M, Veldhoen S, Gratopp A, Mall MA, Eggert A, Deubzer HE. Management of Busulfan-Induced Lung Injury in Pediatric Patients with High-Risk Neuroblastoma. J Clin Med 2024; 13:5995. [PMID: 39408056 PMCID: PMC11477708 DOI: 10.3390/jcm13195995] [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: 09/11/2024] [Revised: 09/29/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Integrating the cytotoxic drug busulfan into a high-dose chemotherapy regimen prior to autologous hematopoietic stem cell rescue in patients with high-risk neuroblastoma has improved the survival of children battling this deadly disease. Busulfan-induced toxicities can, however, be severe. Here, we describe the diagnosis and successful treatment of acute pulmonary injury by total-body-weight-adjusted busulfan therapy in two children with high-risk neuroblastoma. Case series: Patient 1 developed life-threatening biphasic acute respiratory failure on days +60 and +100 after busulfan therapy, requiring intubation and invasive mechanical ventilation. Despite intensive anti-inflammatory and immunomodulatory therapy, including systemic corticosteroids, topical inhalation regimens, azithromycin, nintedanib and extracorporal photopheresis, patient 1 required extended intensive care measures and non-invasive respiratory support for a total of 20 months. High-resolution computed tomography showed diffuse intra-alveolar and interstitial patterns. Patient 2 developed partial respiratory failure with insufficient oxygen saturation and dyspnea on day +52 after busulfan therapy. Symptoms were resolved after 6 months of systemic corticosteroids, topical inhalation regimens and azithromycin. High-resolution computed tomography showed atypical pneumonic changes with ground-glass opacities. While both patients fully recovered without evidence of pulmonary fibrosis, cancer therapy had to be paused and then modified until full recovery from busulfan-induced lung injury. Conclusions: Busulfan-induced lung injury requires prompt diagnosis and intervention. Symptoms and signs are nonspecific and difficult to differentiate from other causes. Therapeutic busulfan drug level monitoring and the identification of patients at risk for drug overdosing through promoter polymorphisms in the glutathione S-transferase alpha 1 gene encoding the main enzyme in busulfan metabolism are expected to reduce the risk of busulfan-induced toxicities.
Collapse
Affiliation(s)
- Sveva Castelli
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Anne Thorwarth
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Claudia van Schewick
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Anke Wendt
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
| | - Annabell Szymansky
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Marco Lodrini
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Simon Veldhoen
- Department of Pediatric Radiology, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Alexander Gratopp
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Marcus A. Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité–Universitätsmedizin Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Lung Research (DLZ), Associated Partner Site Berlin, 89337 Berlin, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, 89337 Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
| | - Hedwig E. Deubzer
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Campus Virchow Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany; (S.C.); (A.S.); (M.L.)
- German Cancer Consortium (DKTK), Partner Site Berlin and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Straße 2, 10178 Berlin, Germany
- Experimental and Clinical Research Center (ECRC) of Charité and Max-Delbrück-Center of Molecular Medicine in the Helmholtz Association, Lindenberger Weg 80, 13125 Berlin, Germany
- Max-Delbrück Center of Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125 Berlin, Germany
| |
Collapse
|
29
|
Fischer T, Dietlein F, Bongartz D, Klehr M, Zimmermanns B, Schmidt M, Mohr A, Mohr F, Sudbrock F, Krapf P, Drzezga A, Dietlein M, Schomäcker K. Tumoricidal Activity and Side Effects of Radiolabeled Anti-NCAM [ 131I]-Iodine-ERIC1 in Neuroblastoma-Bearing Mice. Int J Mol Sci 2024; 25:10737. [PMID: 39409066 PMCID: PMC11476365 DOI: 10.3390/ijms251910737] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Preliminary studies on a radioactive antibody against the neural cell adhesion molecule (NCAM) demonstrated a significant accumulation of [131I]I-ERIC1 in neuroblastoma tumor cells in mice. This study aims to validate the therapeutic efficacy and potential adverse effects of these radioactive immunoconjugates (RICs) in neuroblastoma-bearing mice. To determine the highest tolerated dose, healthy SCID mice received 1 to 22 MBq of [131I]I-ERIC1, with the survival time measured. Tumor response was evaluated by administering 0.8 to 22 MBq of [131I]I-ERIC1 to neuroblastoma-bearing mice and assessing tumor size and systemic toxicity through body weight, blood counts, and survival. It was observed that doses up to approximately 3 MBq per animal (150 MBq/kg) were well tolerated, whereas higher doses resulted in systemic toxicity and death. The neuroblastomas exhibited a dose-dependent response, with optimal therapeutic efficacy achieved at 1.8-2.5 MBq per animal (90-125 MBq/kg), significantly extending survival by a factor of five. The antibody ERIC1 is a promising vehicle for the transport of beta emitters into NCAM-positive tumor tissue. An optimal dosage of the [131I]I-ERIC1 antibody can be established with a balance of tumor-static effects and adverse effects, resulting in a marked extension of survival time.
Collapse
Affiliation(s)
- Thomas Fischer
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
| | - Felix Dietlein
- Computational Health Informatics Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Detlev Bongartz
- Department of Pediatric Oncology and Hematology, Center for Integrated Oncology, University of Cologne, 50937 Cologne, Germany;
| | - Martin Klehr
- Department of Pain Management, Helios Hospital, 47805 Krefeld, Germany;
| | - Beate Zimmermanns
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
| | - Matthias Schmidt
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
| | - Angela Mohr
- School of Mathematics and Science, Inorganic Chemistry, University Wuppertal, 42119 Wuppertal, Germany; (A.M.); (F.M.)
| | - Fabian Mohr
- School of Mathematics and Science, Inorganic Chemistry, University Wuppertal, 42119 Wuppertal, Germany; (A.M.); (F.M.)
| | - Ferdinand Sudbrock
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
| | - Philipp Krapf
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428 Jülich, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Wilhelm-Johnen-Straße, 52428 Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
| | - Klaus Schomäcker
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; (T.F.); (B.Z.); (M.S.); (F.S.); (P.K.); (A.D.); (M.D.)
| |
Collapse
|
30
|
Park SWS, Fransson S, Sundquist F, Nilsson JN, Grybäck P, Wessman S, Strömgren J, Djos A, Fagman H, Sjögren H, Georgantzi K, Herold N, Kogner P, Granberg D, Gaze MN, Martinsson T, Karlsson K, Stenman JJE. Heterogeneous SSTR2 target expression and a novel KIAA1549:: BRAF fusion clone in a progressive metastatic lesion following 177Lutetium-DOTATATE molecular radiotherapy in neuroblastoma: a case report. Front Oncol 2024; 14:1408729. [PMID: 39324010 PMCID: PMC11422106 DOI: 10.3389/fonc.2024.1408729] [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: 03/28/2024] [Accepted: 06/20/2024] [Indexed: 09/27/2024] Open
Abstract
In this case report, we present the treatment outcomes of the first patient enrolled in the LuDO-N trial. The patient is a 21-month-old girl diagnosed with high-risk neuroblastoma (NB) and widespread skeletal metastasis. The patient initially underwent first-line therapy according to SIOPEN HRNBL-1 but was switched to second-line treatments due to disease progression, and she was finally screened for enrollment in the LuDO-N trial due to refractory disease. Upon enrollment, the patient received two rounds of the radiolabeled somatostatin analogue lutetium-177 octreotate (177Lu-DOTATATE), which was well tolerated. A dosimetry analysis revealed a heterogeneous uptake across tumor lesions, resulting in a significant absorbed dose of 54 Gy in the primary tumor, but only 2 Gy at one of the metastatic sites in the distal femur. While the initial treatment response showed disease stabilization, the distal femoral metastasis continued to progress, leading to the eventual death of the patient. A tissue analysis of the biopsies collected throughout the course of the disease revealed heterogeneous drug target expression of somatostatin receptor 2 (SSTR2) across and within tumor lesions. Furthermore, genomic profiling revealed a novel KIAA1549::BRAF fusion oncogene amplification in the distal femoral metastasis at recurrence that might be related with resistance to radiation, possibly through the downregulation of SSTR2. This case report demonstrates a mixed response to molecular radiotherapy (MRT) with 177Lu-DOTATATE. The observed variation in SSTR2 expression between tumor lesions suggests that heterogeneous target expression may have been the reason for treatment failure in this patient's case. Further investigation within the LuDO-N trial will give a more comprehensive understanding of the correlation between SSTR2 expression levels and treatment outcomes, which will be important to advance treatment strategies based on MRT for children with high-risk NB.
Collapse
Affiliation(s)
- Se Whee Sammy Park
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Sundquist
- Department of Urology, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Joachim N Nilsson
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Per Grybäck
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Wessman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Jacob Strömgren
- Department of Pediatric Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Djos
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Fagman
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helene Sjögren
- Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kleopatra Georgantzi
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Nikolas Herold
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Kogner
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Dan Granberg
- Department of Breast, Endocrine Tumors and Sarcomas, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mark N Gaze
- Department of Oncology, University College London Hospitals National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Tommy Martinsson
- Department of Laboratory Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kasper Karlsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jakob J E Stenman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
31
|
Mohseni R, Mahdavi Sharif P, Behfar M, Shojaei S, Shoae-Hassani A, Jafari L, Khosravi A, Nikfetrat Z, Hamidieh AA. Phase I study of safety and efficacy of allogeneic natural killer cell therapy in relapsed/refractory neuroblastomas post autologous hematopoietic stem cell transplantation. Sci Rep 2024; 14:20971. [PMID: 39251669 PMCID: PMC11385932 DOI: 10.1038/s41598-024-70958-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 08/22/2024] [Indexed: 09/11/2024] Open
Abstract
Despite low incidence, neuroblastoma, an immunologically cold tumor, is the most common extracranial solid neoplasm in pediatrics. In relapsed/refractory cases, the benefits of autologous hematopoietic stem cell transplantation (auto-HSCT) and other therapies are limited. Natural killer (NK) cells apply cytotoxicity against tumor cells independently of antigen-presenting cells and the adaptive immune system. The primary endpoint of this trial was to assess the safety of the injection of allogenic, ex vivo-expanded and primed NK cells in relapsed/refractory neuroblastoma patients after auto-HSCT. The secondary endpoint included the efficacy of this intervention in controlling tumors. NK cells were isolated and primed ex vivo (by adding interleukin [IL]-2, IL-15, and IL-21) in a GMP-compliant CliniMACS system and administered to four patients with relapsed/refractory MYCN-positive neuroblastoma. NK cell injections (1 and 5 × 107 cells/kg in the first and second injections, respectively) were safe, and no acute or sub-acute adverse events were observed. During the follow-up period, one complete response (CR) and one partial response (PR) were observed, while two cases exhibited progressive disease (PD). In follow-up evaluations, two died due to disease progression, including the case with a PR. The patient with CR had regular growth at the 31-month follow-up, and another patient with PD is still alive and receiving chemotherapies 20 months after therapy. This therapy is an appealing and feasible approach for managing refractory neuroblastomas post-HSCT. Further studies are needed to explore its efficacy with higher doses and more frequent administrations for high-risk neuroblastomas and other immunologically cold tumors.Trial registration number: irct.behdasht.gov.ir (Iranian Registry of Clinical Trials, No. IRCT20201202049568N1).
Collapse
Affiliation(s)
- Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran
| | - Pouya Mahdavi Sharif
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran
| | - Sahar Shojaei
- National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Alireza Shoae-Hassani
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Jafari
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran
| | - Abbas Khosravi
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran
| | - Zeynab Nikfetrat
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, 14194, Iran.
| |
Collapse
|
32
|
Seo ES, Lee JW, Cho HW, Ju HY, Cho YS, Lee S, Moon SH, Yoo KH, Lim DH, Sung KW. Response-adapted consolidation therapy strategy for patients with metastatic high-risk neuroblastoma: Results of the SMC NB-2014 study. Pediatr Blood Cancer 2024; 71:e31173. [PMID: 38965702 DOI: 10.1002/pbc.31173] [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: 02/13/2024] [Revised: 05/28/2024] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Tandem high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT) and incorporation of 131I-metaiodobenzylguanidine (131I-MIBG) treatment have shown positive outcomes in high-risk neuroblastoma. However, more optimized treatment strategies are still needed. PROCEDURE The NB-2014 study was a nonrandomized, prospective trial that examined survival outcomes in metastatic high-risk neuroblastoma patients using response-adapted consolidation therapy. We used post-induction residual 123I-MIBG status at metastatic sites as a treatment response marker. Patients achieving complete resolution of MIBG uptake at metastatic sites underwent a reduced first HDCT/auto-SCT with a 20% dose reduction in HDCT. After the first HDCT/auto-SCT, patients with remaining MIBG uptake received dose-escalated (18 mCi/kg) 131I-MIBG treatment. In contrast, those with complete resolution of MIBG at metastatic sites received a standard dose (12 mCi/kg) of 131I-MIBG. We compared survival and toxicity outcomes with a historical control group from the NB-2009. RESULTS Of 65 patients treated, 63% achieved complete resolution of MIBG uptake at metastatic sites following induction chemotherapy, while 29% of patients still had MIBG uptake at metastatic sites after the first HDCT/auto-SCT. The 3-year event-free survival (EFS) and overall survival (OS) rates were 68.2% ± 6.0% and 86.5% ± 4.5%, respectively. Compared to NB-2009, EFS was similar (p = .855); however, NB-2014 had a higher OS (p = .031), a lower cumulative incidence of treatment-related mortality (p = .036), and fewer acute and late toxicities. CONCLUSIONS Our results suggest that response-adaptive consolidation therapy based on chemotherapy response at metastatic sites facilitates better treatment tailoring, and appears promising for patients with metastatic high-risk neuroblastoma.
Collapse
Affiliation(s)
- Eun Seop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sanghoon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seung Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Do Hoon Lim
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
33
|
Ek T, Ibrahim RR, Vogt H, Georgantzi K, Träger C, Gaarder J, Djos A, Rahmqvist I, Mellström E, Pujol-Calderón F, Vannas C, Hansson L, Fagman H, Treis D, Fransson S, Österlund T, Chuang TP, Verhoeven BM, Ståhlberg A, Palmer RH, Hallberg B, Martinsson T, Kogner P, Dalin M. Long-Lasting Response to Lorlatinib in Patients with ALK-Driven Relapsed or Refractory Neuroblastoma Monitored with Circulating Tumor DNA Analysis. CANCER RESEARCH COMMUNICATIONS 2024; 4:2553-2564. [PMID: 39177282 PMCID: PMC11440348 DOI: 10.1158/2767-9764.crc-24-0338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
Patients with anaplastic lymphoma kinase (ALK)-driven neuroblastoma may respond to tyrosine kinase inhibitors, but resistance to treatment occurs and methods currently used for detection of residual disease have limited sensitivity. Here, we present a national unselected cohort of five patients with relapsed or refractory ALK-driven neuroblastoma treated with lorlatinib as monotherapy and test the potential of targeted circulating tumor DNA (ctDNA) analysis as a guide for treatment decisions in these patients. We developed a sequencing panel for ultrasensitive detection of ALK mutations associated with neuroblastoma or resistance to tyrosine kinase inhibitors and used it for ctDNA analysis in 83 plasma samples collected longitudinally from the four patients who harbored somatic ALK mutations. All four patients with ALK p.R1275Q experienced major responses and were alive 35 to 61 months after starting lorlatinib. A fifth patient with ALK p.F1174L initially had a partial response but relapsed after 10 months of treatment. In all cases, ctDNA was detected at the start of lorlatinib single-agent treatment and declined gradually, correlating with clinical responses. In the two patients exhibiting relapse, ctDNA increased 9 and 3 months, respectively, before clinical detection of disease progression. In one patient harboring HRAS p.Q61L in the relapsed tumor, retrospective ctDNA analysis showed that the mutation appeared de novo after 8 months of lorlatinib treatment. We conclude that some patients with relapsed or refractory high-risk neuroblastoma show durable responses to lorlatinib as monotherapy, and targeted ctDNA analysis is effective for evaluation of treatment and early detection of relapse in ALK-driven neuroblastoma. SIGNIFICANCE We present five patients with ALK-driven relapsed or refractory neuroblastoma treated with lorlatinib as monotherapy. All patients responded to treatment, and four of them were alive after 3 to 5 years of follow-up. We performed longitudinal ctDNA analysis with ultra-deep sequencing of the ALK tyrosine kinase domain. We conclude that ctDNA analysis may guide treatment decisions in ALK-driven neuroblastoma, also when the disease is undetectable using standard clinical methods.
Collapse
Affiliation(s)
- Torben Ek
- Children’s Cancer Centre, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Raghda R. Ibrahim
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Hartmut Vogt
- Department of Biomedical and Clinical Sciences, Crown Princess Victoria Children’s Hospital, and Division of Children’s and Women’s Health, Linköping University, Linköping, Sweden.
| | - Kleopatra Georgantzi
- Department of Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden.
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Catarina Träger
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
- Department of Pediatric Hematology and Oncology, Academic Children’s Hospital, Uppsala, Sweden.
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden.
| | - Jennie Gaarder
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Ida Rahmqvist
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Elisabeth Mellström
- Children’s Cancer Centre, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Fani Pujol-Calderón
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Christoffer Vannas
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Lina Hansson
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Henrik Fagman
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Pathology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Diana Treis
- Department of Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden.
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
| | - Tobias Österlund
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Tzu-Po Chuang
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Bronte Manouk Verhoeven
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Anders Ståhlberg
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Ruth H. Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Per Kogner
- Department of Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden.
- Childhood Cancer Research Unit, Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden.
| | - Martin Dalin
- Children’s Cancer Centre, Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden.
- Department of Pediatrics, Sahlgrenska Center for Cancer Research, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
34
|
Foster JH, Reid JM, Minard C, Woodfield S, Denic KZ, Isikwei E, Voss SD, Nelson M, Liu X, Berg SL, Fox E, Weigel BJ. Phase 1 study of NEDD8 activating enzyme inhibitor pevonedistat in combination with chemotherapy in pediatric patients with recurrent or refractory solid tumors (ADVL1615). Eur J Cancer 2024; 209:114241. [PMID: 39096851 PMCID: PMC11392690 DOI: 10.1016/j.ejca.2024.114241] [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: 04/16/2024] [Revised: 06/24/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024]
Abstract
PURPOSE The objective of this study was to determine the recommended Phase 2 dose (RP2D) of pevonedistat, a first in class inhibitor of NEDD8 activating enzyme, in combination with irinotecan (IRN) and temozolomide (TMZ) in children with cancer. METHODS This Phase 1 study used a rolling 6 design to evaluate escalating doses of pevonedistat in combination with standard doses of IRN and TMZ in pediatric patients with recurrent/refractory solid or CNS tumors. During cycle 1, pevonedistat was administered intravenously on days 1, 8, 10, and 12, with IRN (IV, 50 mg/m2) and TMZ (orally, 100 mg/m2), on days 8-12 of a 28-day cycle. In subsequent cycles, pevonedistat was administered on days 1, 3, and 5, with IRN/TMZ on days 1-5 of a 21-day cycle. RESULTS Thirty patients enrolled; all were eligible and evaluable for toxicity. Six patients each enrolled on pevonedistat dose levels (DL) 1 (15 mg/m2), 2 (20 mg/m2), 3 (25 mg/m2) and 4 (35 mg/m2) as well as an expanded pharmacokinetic (PK) cohort at DL4. The maximum tolerated dose (MTD) was not exceeded. 2/12 (17 %) patients treated at the RP2D (35 mg/m2) experienced a cycle 1 dose limiting toxicity (DLT). IRN is unlikely to affect the pharmacokinetics of pevonedistat. Two patients had a partial response and 6 patients had prolonged stable disease (> 6 cycles). CONCLUSIONS Pevonedistat in combination with IRN/TMZ is well tolerated in children with solid or CNS tumors. The RP2D of pevonedistat is 35 mg/m2 on days 1, 3, 5 in combination with IRN/TMZ.
Collapse
Affiliation(s)
- Jennifer H Foster
- Texas Children's Hospital, Baylor College of Medicine, Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA.
| | | | - Charles Minard
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Sarah Woodfield
- Texas Children's Hospital, Baylor College of Medicine, Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | | | | | - Stephan D Voss
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marvin Nelson
- Children's Hospital Los Angeles, Radiology, Keck USC School of Medicine, Los Angeles, CA, USA
| | - Xiaowei Liu
- Children's Oncology Group, Monrovia, CA, USA
| | - Stacey L Berg
- Texas Children's Hospital, Baylor College of Medicine, Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Elizabeth Fox
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Brenda J Weigel
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
35
|
Cupit-Link M, Hagiwara K, Nagy M, Koo SC, Orr BA, Ruppin E, Easton J, Zhang J, Federico SM. Response to PARP Inhibition in BARD1-Mutated Refractory Neuroblastoma. N Engl J Med 2024; 391:659-661. [PMID: 39141861 PMCID: PMC11328958 DOI: 10.1056/nejmc2403316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Affiliation(s)
| | | | | | - Selene C Koo
- St. Jude Children's Research Hospital, Memphis, TN
| | - Brent A Orr
- St. Jude Children's Research Hospital, Memphis, TN
| | | | - John Easton
- St. Jude Children's Research Hospital, Memphis, TN
| | | | | |
Collapse
|
36
|
Bobin C, Iddir Y, Butterworth C, Masliah-Planchon J, Saint-Charles A, Bellini A, Bhalshankar J, Pierron G, Combaret V, Attignon V, André N, Corradini N, Dumont B, Mansuy L, Khanfar C, Klein S, Briandet C, Plantaz D, Millot F, Thouvenin S, Aerts I, Ndounga-Diakou LA, Laghouati S, Abbou S, Jehanno N, Tissot H, Renault S, Baulande S, Raynal V, Bozec L, Bieche I, Delattre O, Berlanga P, Schleiermacher G. Sequential Analysis of cfDNA Reveals Clonal Evolution in Patients with Neuroblastoma Receiving ALK-Targeted Therapy. Clin Cancer Res 2024; 30:3316-3328. [PMID: 38787533 PMCID: PMC11292203 DOI: 10.1158/1078-0432.ccr-24-0753] [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: 03/06/2024] [Revised: 04/09/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE The study of cell-free DNA (cfDNA) enables sequential analysis of tumor cell-specific genetic alterations in patients with neuroblastoma. EXPERIMENTAL DESIGN Eighteen patients with relapsing neuroblastoma having received lorlatinib, a third-generation ALK inhibitor, were identified (SACHA national registry and/or in the institution). cfDNA was analyzed at relapse for nine patients and sequentially for five patients (blood/bone marrow plasma) by performing whole-genome sequencing library construction followed by ALK-targeted ddPCR of the hotspot mutations [F1174L, R1275Q, and I1170N; variant allele fraction (VAF) detection limit 0.1%] and whole-exome sequencing (WES) to evaluate disease burden and clonal evolution, following comparison with tumor/germline WES. RESULTS Overall response rate to lorlatinib was 33% (CI, 13%-59%), with response observed in 6/10 cases without versus 0/8 cases with MYCN amplification (MNA). ALK VAFs correlated with the overall clinical disease status, with a VAF < 0.1% in clinical remission, versus higher VAFs (>30%) at progression. Importantly, sequential ALK ddPCR detected relapse earlier than clinical imaging. cfDNA WES revealed new SNVs, not seen in the primary tumor, in all instances of disease progression after lorlatinib treatment, indicating clonal evolution, including alterations in genes linked to tumor aggressivity (TP53) or novel targets (EGFR). Gene pathway analysis revealed an enrichment for genes targeting cell differentiation in emerging clones, and cell adhesion in persistent clones. Evidence of clonal hematopoiesis could be observed in follow-up samples. CONCLUSIONS We demonstrate the clinical utility of combining ALK cfDNA ddPCR for disease monitoring and cfDNA WES for the study of clonal evolution and resistance mechanisms in patients with neuroblastoma receiving ALK-targeted therapy.
Collapse
Affiliation(s)
- Charles Bobin
- SiRIC RTOP (Recherche Translationelle en Oncologie Pédiatrique), Translational Research Department, Institut Curie Research Center, PSL Research University, Institut Curie, Paris, France.
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
| | - Yasmine Iddir
- SiRIC RTOP (Recherche Translationelle en Oncologie Pédiatrique), Translational Research Department, Institut Curie Research Center, PSL Research University, Institut Curie, Paris, France.
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
| | - Charlotte Butterworth
- SiRIC RTOP (Recherche Translationelle en Oncologie Pédiatrique), Translational Research Department, Institut Curie Research Center, PSL Research University, Institut Curie, Paris, France.
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
| | | | - Alexandra Saint-Charles
- SiRIC RTOP (Recherche Translationelle en Oncologie Pédiatrique), Translational Research Department, Institut Curie Research Center, PSL Research University, Institut Curie, Paris, France.
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
| | - Angela Bellini
- SiRIC RTOP (Recherche Translationelle en Oncologie Pédiatrique), Translational Research Department, Institut Curie Research Center, PSL Research University, Institut Curie, Paris, France.
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
| | - Jaydutt Bhalshankar
- SiRIC RTOP (Recherche Translationelle en Oncologie Pédiatrique), Translational Research Department, Institut Curie Research Center, PSL Research University, Institut Curie, Paris, France.
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
| | | | - Valérie Combaret
- Laboratoire de Recherche Translationnelle, Centre Léon-Bérard, Lyon, France.
| | - Valéry Attignon
- Laboratoire de Recherche Translationnelle, Centre Léon-Bérard, Lyon, France.
| | - Nicolas André
- Marseille-La Timone University Hospital, Oncologie Pédiatrique, Marseille, France.
- CRCM INSERM U1068 REMAP4KIDS, Aix Marseille University, Marseille, France.
| | - Nadège Corradini
- Department of Pediatric Oncology, Institute for Paediatric Haematology and Oncology, Léon Bérard Center, Lyon, France.
| | - Benoit Dumont
- Department of Pediatric Oncology, Institute for Paediatric Haematology and Oncology, Léon Bérard Center, Lyon, France.
| | - Ludovic Mansuy
- Service d’oncologie Pédiatrique du CHRU de Nancy, Hôpital d’enfants, Vandoeuvre, France.
| | - Camille Khanfar
- Department of Pediatric Oncology, CHU Amiens Picardie, Amiens, France.
| | - Sebastien Klein
- Pediatric Oncology and Hematology, CHU Jean-Minjoz, Besançon, France.
| | | | - Dominique Plantaz
- Department of Pediatric Onco-Immuno-Hematology, Grenoble Alpes University Hospital, Grenoble, France.
| | - Frederic Millot
- Department of Paediatric Haematology and Oncology, Centre Hospitalo-Universitaire de Poitiers, Poitiers, France.
| | - Sandrine Thouvenin
- Department of Pediatric Hematology-Oncology, University Hospital St Etienne, St Etienne, France.
| | - Isabelle Aerts
- SIREDO Integrated Pediatric Oncology Center, Institut Curie, Paris, France.
| | - Lee Aymar Ndounga-Diakou
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
| | - Salim Laghouati
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
| | - Samuel Abbou
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
| | - Nina Jehanno
- Department of Nuclear Medicine, Institut Curie, Paris, France.
| | - Hubert Tissot
- Department of Nuclear Medicine, Institut Curie, Paris, France.
| | - Shufang Renault
- Circulating Tumor Biomarkers Laboratory, Inserm CIC-BT 1428, Department of Translational Research, Institut Curie, Paris, France.
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Research Center, Institut Curie, Paris, France.
| | - Virginie Raynal
- Institut Curie Genomics of Excellence (ICGex) Platform, Research Center, Institut Curie, Paris, France.
| | - Laurence Bozec
- Department of Medical Oncology, Institut Curie, Saint-Cloud, France.
| | - Ivan Bieche
- Pharmacogenomics Unit, Institut Curie, Paris, France.
| | - Olivier Delattre
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
- Somatic Genetics Unit, Institut Curie, Paris, France.
| | - Pablo Berlanga
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
| | - Gudrun Schleiermacher
- SiRIC RTOP (Recherche Translationelle en Oncologie Pédiatrique), Translational Research Department, Institut Curie Research Center, PSL Research University, Institut Curie, Paris, France.
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France.
- SIREDO Integrated Pediatric Oncology Center, Institut Curie, Paris, France.
| |
Collapse
|
37
|
Winnicka D, Skowera P, Stelmach M, Styka B, Lejman M. Application of the FISH method and high-density SNP arrays to assess genetic changes in neuroblastoma-research by one institute. Acta Biochim Pol 2024; 71:12821. [PMID: 39049899 PMCID: PMC11267511 DOI: 10.3389/abp.2024.12821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
Neuroblastoma is the most common extracranial solid tumor in children. Amplification of the MYCN gene has been observed in approximately 20%-30% of tumors. It is strongly correlated with advanced-stage disease, rapid tumor progression, resistance to chemotherapy and poor outcomes independent of patient age and stage of advanced disease. MYCN amplification identifies high-risk patients. To assess neuroblastoma tumors with MYCN amplification we used paraffin-embedded tissue sections in 57 patients and intraoperative tumor imprints in 10 patients by fluorescence in situ hybridization (FISH). Positive results for MYCN amplification have been observed in twelve patients' paraffin-embedded tissue sections and in three patients' intraoperative tumor imprints, which represents 22.4% of all patients tested in the analysis. Fluorescence in situ hybridization is a highly sensitive and useful technique for detecting MYCN amplification on paraffin-embedded tissue sections of neuroblastoma tumors and intraoperative tumor imprints thus facilitating therapeutic decisions based on the presence or absence of this important biologic marker. The presence of structural changes, regardless of MYCN gene amplification status, influences the clinical behavior of neuroblastoma. High-Density SNP Arrays have emerged as the perfect tools for detecting these changes due to their exceptional accuracy, sensitivity and ability to analyze copy number and allele information. Consequently, they are proven to be highly valuable in the genomic diagnosis of immature neuroectodermal tumors.
Collapse
Affiliation(s)
| | | | - Magdalena Stelmach
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Lublin, Poland
| | | | | |
Collapse
|
38
|
Corbacioglu S, Lode H, Ellinger S, Zeman F, Suttorp M, Escherich G, Bochennek K, Gruhn B, Lang P, Rohde M, Debatin KM, Steinbach D, Beilken A, Ladenstein R, Spachtholz R, Heiss P, Hellwig D, Tröger A, Koller M, Menhart K, Riemenschneider MJ, Zoubaa S, Kietz S, Jakob M, Sommer G, Heise T, Hundsdörfer P, Kühnle I, Dilloo D, Schönberger S, Schwabe G, von Luettichau I, Graf N, Schlegel PG, Frühwald M, Jorch N, Paulussen M, Schneider DT, Metzler M, Leipold A, Nathrath M, Imschweiler T, Christiansen H, Schmid I, Crazzolara R, Niktoreh N, Cario G, Faber J, Demmert M, Babor F, Fröhlich B, Bielack S, Bernig T, Greil J, Eggert A, Simon T, Foell J. Irinotecan and temozolomide in combination with dasatinib and rapamycin versus irinotecan and temozolomide for patients with relapsed or refractory neuroblastoma (RIST-rNB-2011): a multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol 2024; 25:922-932. [PMID: 38936379 DOI: 10.1016/s1470-2045(24)00202-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Neuroblastoma is the most common extracranial solid tumour in children. Relapsed or refractory neuroblastoma is associated with a poor outcome. We assessed the combination of irinotecan-temozolomide and dasatinib-rapamycin (RIST) in patients with relapsed or refractory neuroblastoma. METHODS The multicentre, open-label, randomised, controlled, phase 2, RIST-rNB-2011 trial recruited from 40 paediatric oncology centres in Germany and Austria. Patients aged 1-25 years with high-risk relapsed (defined as recurrence of all stage IV and MYCN amplification stages, after response to treatment) or refractory (progressive disease during primary treatment) neuroblastoma, with Lansky and Karnofsky performance status at least 50%, were assigned (1:1) to RIST (RIST group) or irinotecan-temozolomide (control group) by block randomisation, stratified by MYCN status. We compared RIST (oral rapamycin [loading 3 mg/m2 on day 1, maintenance 1 mg/m2 on days 2-4] and oral dasatinib [2 mg/kg per day] for 4 days with 3 days off, followed by intravenous irinotecan [50 mg/m2 per day] and oral temozolomide [150 mg/m2 per day] for 5 days with 2 days off; one course each of rapamycin-dasatinib and irinotecan-temozolomide for four cycles over 8 weeks, then two courses of rapamycin-dasatinib followed by one course of irinotecan-temozolomide for 12 weeks) with irinotecan-temozolomide alone (with identical dosing as experimental group). The primary endpoint of progression-free survival was analysed in all eligible patients who received at least one course of therapy. The safety population consisted of all patients who received at least one course of therapy and had at least one post-baseline safety assessment. This trial is registered at ClinicalTrials.gov, NCT01467986, and is closed to accrual. FINDINGS Between Aug 26, 2013, and Sept 21, 2020, 129 patients were randomly assigned to the RIST group (n=63) or control group (n=66). Median age was 5·4 years (IQR 3·7-8·1). 124 patients (78 [63%] male and 46 [37%] female) were included in the efficacy analysis. At a median follow-up of 72 months (IQR 31-88), the median progression-free survival was 11 months (95% CI 7-17) in the RIST group and 5 months (2-8) in the control group (hazard ratio 0·62, one-sided 90% CI 0·81; p=0·019). Median progression-free survival in patients with amplified MYCN (n=48) was 6 months (95% CI 4-24) in the RIST group versus 2 months (2-5) in the control group (HR 0·45 [95% CI 0·24-0·84], p=0·012); median progression-free survival in patients without amplified MYCN (n=76) was 14 months (95% CI 9-7) in the RIST group versus 8 months (4-15) in the control group (HR 0·84 [95% CI 0·51-1·38], p=0·49). The most common grade 3 or worse adverse events were neutropenia (54 [81%] of 67 patients given RIST vs 49 [82%] of 60 patients given control), thrombocytopenia (45 [67%] vs 41 [68%]), and anaemia (39 [58%] vs 38 [63%]). Nine serious treatment-related adverse events were reported (five patients given control and four patients given RIST). There were no treatment-related deaths in the control group and one in the RIST group (multiorgan failure). INTERPRETATION RIST-rNB-2011 demonstrated that targeting of MYCN-amplified relapsed or refractory neuroblastoma with a pathway-directed metronomic combination of a multkinase inhibitor and an mTOR inhibitor can improve progression-free survival and overall survival. This exclusive efficacy in MYCN-amplified, relapsed neuroblastoma warrants further investigation in the first-line setting. FUNDING Deutsche Krebshilfe.
Collapse
Affiliation(s)
- Selim Corbacioglu
- University Medical Center Regensburg, Regensburg, Germany; Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation,University Medical Center Regensburg, Regensburg, Germany.
| | - Holger Lode
- University Medicine Greifswald, Greifswald, Germany
| | | | - Florian Zeman
- University Medical Center Regensburg, Regensburg, Germany
| | - Meinolf Suttorp
- Medical Faculty, Technical University Dresden, Dresden, Germany
| | | | | | - Bernd Gruhn
- University Medical Center Jena, Jena, Germany
| | - Peter Lang
- University Medical Center Tuebingen, Tuebingen, Germany
| | - Marius Rohde
- University Medical Center Giessen, Giessen, Germany
| | | | | | | | - Ruth Ladenstein
- St Anna Children's Cancer Research Institute, University Medical Center, Vienna, Austria
| | | | - Peter Heiss
- University Medical Center Regensburg, Regensburg, Germany
| | - Dirk Hellwig
- University Medical Center Regensburg, Regensburg, Germany
| | - Anja Tröger
- University Medical Center Regensburg, Regensburg, Germany
| | - Michael Koller
- University Medical Center Regensburg, Regensburg, Germany
| | - Karin Menhart
- University Medical Center Regensburg, Regensburg, Germany
| | | | - Saida Zoubaa
- University Medical Center Regensburg, Regensburg, Germany
| | - Silke Kietz
- University Medical Center Regensburg, Regensburg, Germany
| | - Marcus Jakob
- University Medical Center Regensburg, Regensburg, Germany
| | - Gunhild Sommer
- University Medical Center Regensburg, Regensburg, Germany
| | - Tilman Heise
- University Medical Center Regensburg, Regensburg, Germany
| | | | - Ingrid Kühnle
- University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | - Norbert Jorch
- University Medical Center Bielefeld, Bielefeld, Germany
| | | | | | | | | | | | | | | | - Irene Schmid
- Ludwig Maximilians University Munich, Munich, Germany
| | | | | | | | - Joerg Faber
- University Medical Center Mainz, Mainz, Germany
| | | | - Florian Babor
- University Medical Center Düsseldorf, Düsseldorf, Germany
| | | | | | | | - Johann Greil
- University Medical Center Heidelberg, Heidelberg, Germany
| | | | | | - Juergen Foell
- University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
39
|
de Faria LL, Ponich Clementino C, Véras FASE, Khalil DDC, Otto DY, Oranges Filho M, Suzuki L, Bedoya MA. Staging and Restaging Pediatric Abdominal and Pelvic Tumors: A Practical Guide. Radiographics 2024; 44:e230175. [PMID: 38722785 DOI: 10.1148/rg.230175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The most common abdominal malignancies diagnosed in the pediatric population include neuroblastoma, Wilms tumor, hepatoblastoma, lymphoma, germ cell tumor, and rhabdomyosarcoma. There are distinctive imaging findings and patterns of spread for each of these tumors that radiologists must know for diagnosis and staging and for monitoring the patient's response to treatment. The multidisciplinary treatment group that includes oncologists, surgeons, and radiation oncologists relies heavily on imaging evaluation to identify the best treatment course and prognostication of imaging findings, such as the image-defined risk factors for neuroblastomas, the PRETreatment EXtent of Disease staging system for hepatoblastoma, and the Ann Arbor staging system for lymphomas. It is imperative for radiologists to be able to correctly indicate the best imaging methods for diagnosis, staging, and restaging of each of these most prevalent tumors to avoid inconclusive or unnecessary examinations. The authors review in a practical manner the most updated key points in diagnosing and staging disease and assessing response to treatment of the most common pediatric abdominal tumors. ©RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Luisa Leitão de Faria
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Carolina Ponich Clementino
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Felippe Augusto Silvestre E Véras
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Douglas da Cunha Khalil
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Deborah Yukiko Otto
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Marcelo Oranges Filho
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - Lisa Suzuki
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| | - M Alejandra Bedoya
- From the Radiology Department, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Rua Dr Ovídio Pires de Campos, 225 Cerqueira César, São Paulo, SP 36070-460, Brazil (L.L.d.F., C.P.C., F.A.S.e.V., D.d.C.K., D.Y.O., M.O.F., L.S.); and Department of Radiology, Boston Childrens Hospital, Harvard Medical School, Boston, Mass (M.A.B.)
| |
Collapse
|
40
|
Zhao JD, Lu XY, Chen TP, Duan XL, Zuo W, Sai K, Zhu LR, Gao Q. Development and validation of a novel nomogram for predicting overall survival patients with neuroblastoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108321. [PMID: 38598875 DOI: 10.1016/j.ejso.2024.108321] [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: 12/13/2023] [Revised: 03/26/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
PURPOSE The aim of this study was to develop a nomogram specially for predicting overall survival (OS) for Chinese patients with neuroblastoma (NB). METHODS Patients with pathologically confirmed NB who were newly diagnosed and received treatments at our hospital from October 2013 to October 2021 were retrospectively reviewed. The nomogram for OS were built based on Cox regression analysis. The validation of the prognostic model was evaluated by concordance index (C-index), calibration curves, and decision curve analyses (DCAs). RESULTS A total of 254 patients with NB were included in this study. They were randomly divided into a training cohort (n = 178) and a validation cohort (n = 76) at a ratio of 7:3. Multivariate analyses revealed that prognostic variables significantly related to the OS were age at diagnosis, bone metastasis, hepatic metastasis, INSS stage, MYCN status and DNA ploidy. The nomogram was constructed based on above 6 factors. The C-index values of the nomogram for predicting 3-year and 5-year OS were 0.926 and 0.964, respectively. The calibration curves of the nomogram showed good consistency between nomogram prediction and actual survival. The DCAs showed great clinical usefulness of the nomograms. Furthermore, patients with low-risk identified by our nomogram had much higher OS than those with high-risk (p < 0.001). CONCLUSION The nomogram we constructed exhibited good predictive performance and could be used to assist clinicians in their decision-making process.
Collapse
Affiliation(s)
- Jin-du Zhao
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Xian-Ying Lu
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Tian-Ping Chen
- Department of Hematology and Oncology, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Xian-Lun Duan
- Department of Thoracic Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Wei Zuo
- Department of Neonatal Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Kai Sai
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Li-Ran Zhu
- Anhui Institute of Pediatric Research, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China
| | - Qun Gao
- Department of Oncology Surgery, Anhui Medical University Children's Medical Center, Anhui Provincial Children's Hospital, Hefei, 230051, Anhui, China.
| |
Collapse
|
41
|
Cheung IY, Mauguen A, Modak S, Basu EM, Feng Y, Kushner BH, Cheung NK. Long Prime-Boost Interval and Heightened Anti-GD2 Antibody Response to Carbohydrate Cancer Vaccine. Vaccines (Basel) 2024; 12:587. [PMID: 38932316 PMCID: PMC11209353 DOI: 10.3390/vaccines12060587] [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: 04/04/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
The carbohydrate ganglioside GD2/GD3 cancer vaccine adjuvanted by β-glucan stimulates anti-GD2 IgG1 antibodies that strongly correlate with improved progression-free survival (PFS) and overall survival (OS) among patients with high-risk neuroblastoma. Thirty-two patients who relapsed on the vaccine (first enrollment) were re-treated on the same vaccine protocol (re-enrollment). Titers during the first enrollment peaked by week 32 at 751 ± 270 ng/mL, which plateaued despite vaccine boosts at 1.2-4.5 month intervals. After a median wash-out interval of 16.1 months from the last vaccine dose during the first enrollment to the first vaccine dose during re-enrollment, the anti-GD2 IgG1 antibody rose to a peak of 4066 ± 813 ng/mL by week 3 following re-enrollment (p < 0.0001 by the Wilcoxon matched-pairs signed-rank test). Yet, these peaks dropped sharply and continually despite repeated boosts at 1.2-4.5 month intervals, before leveling off by week 20 to the first enrollment peak levels. Despite higher antibody titers, patients experienced no pain or neuropathic side effects, which were typically associated with immunotherapy using monoclonal anti-GD2 antibodies. By the Kaplan-Meier method, PFS was estimated to be 51%, and OS was 81%. The association between IgG1 titer during re-enrollment and β-glucan receptor dectin-1 SNP rs3901533 was significant (p = 0.01). A longer prime-boost interval could significantly improve antibody responses in patients treated with ganglioside conjugate cancer vaccines.
Collapse
Affiliation(s)
- Irene Y. Cheung
- Departments of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (S.M.); (E.M.B.); (Y.F.); (B.H.K.); (N.K.C.)
| | - Audrey Mauguen
- Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA;
| | - Shakeel Modak
- Departments of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (S.M.); (E.M.B.); (Y.F.); (B.H.K.); (N.K.C.)
| | - Ellen M. Basu
- Departments of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (S.M.); (E.M.B.); (Y.F.); (B.H.K.); (N.K.C.)
| | - Yi Feng
- Departments of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (S.M.); (E.M.B.); (Y.F.); (B.H.K.); (N.K.C.)
| | - Brian H. Kushner
- Departments of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (S.M.); (E.M.B.); (Y.F.); (B.H.K.); (N.K.C.)
| | - Nai Kong Cheung
- Departments of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; (S.M.); (E.M.B.); (Y.F.); (B.H.K.); (N.K.C.)
| |
Collapse
|
42
|
Deyell RJ, Shen Y, Titmuss E, Dixon K, Williamson LM, Pleasance E, Nelson JMT, Abbasi S, Krzywinski M, Armstrong L, Bonakdar M, Ch'ng C, Chuah E, Dunham C, Fok A, Jones M, Lee AF, Ma Y, Moore RA, Mungall AJ, Mungall KL, Rogers PC, Schrader KA, Virani A, Wee K, Young SS, Zhao Y, Jones SJM, Laskin J, Marra MA, Rassekh SR. Whole genome and transcriptome integrated analyses guide clinical care of pediatric poor prognosis cancers. Nat Commun 2024; 15:4165. [PMID: 38755180 PMCID: PMC11099106 DOI: 10.1038/s41467-024-48363-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
The role for routine whole genome and transcriptome analysis (WGTA) for poor prognosis pediatric cancers remains undetermined. Here, we characterize somatic mutations, structural rearrangements, copy number variants, gene expression, immuno-profiles and germline cancer predisposition variants in children and adolescents with relapsed, refractory or poor prognosis malignancies who underwent somatic WGTA and matched germline sequencing. Seventy-nine participants with a median age at enrollment of 8.8 y (range 6 months to 21.2 y) are included. Germline pathogenic/likely pathogenic variants are identified in 12% of participants, of which 60% were not known prior. Therapeutically actionable variants are identified by targeted gene report and whole genome in 32% and 62% of participants, respectively, and increase to 96% after integrating transcriptome analyses. Thirty-two molecularly informed therapies are pursued in 28 participants with 54% achieving a clinical benefit rate; objective response or stable disease ≥6 months. Integrated WGTA identifies therapeutically actionable variants in almost all tumors and are directly translatable to clinical care of children with poor prognosis cancers.
Collapse
Affiliation(s)
- Rebecca J Deyell
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada.
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Emma Titmuss
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Katherine Dixon
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Erin Pleasance
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Jessica M T Nelson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Sanna Abbasi
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Martin Krzywinski
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Linlea Armstrong
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Melika Bonakdar
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Carolyn Ch'ng
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Chris Dunham
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Alexandra Fok
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Martin Jones
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Anna F Lee
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yussanne Ma
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Paul C Rogers
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada
| | - Kasmintan A Schrader
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Alice Virani
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Kathleen Wee
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Sean S Young
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Cancer Genetics and Genomics Laboratory, Department of Pathology and Laboratory Medicine, BC Cancer, Vancouver, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Shahrad R Rassekh
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
43
|
Raiser P, Schleiermacher G, Gambart M, Dumont B, Defachelles AS, Thebaud E, Tandonnet J, Pasqualini C, Proust S, Entz-Werle N, Aerts I, Ndounga-Diakou LA, Petit A, Puiseux C, Khanfar C, Rouger J, Mansuy L, Benadiba J, Millot F, Pluchart C, Laghouati S, Geoerger B, Vassal G, Valteau-Couanet D, Berlanga P. Chemo-immunotherapy with dinutuximab beta in patients with relapsed/progressive high-risk neuroblastoma: does chemotherapy backbone matter? Eur J Cancer 2024; 202:114001. [PMID: 38489858 DOI: 10.1016/j.ejca.2024.114001] [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: 01/16/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Addition of anti-GD2 antibodies to temozolomide-based chemotherapy has demonstrated increased antitumor activity and progression-free survival in patients with relapsed/progressive high-risk neuroblastoma. However, chemo-immunotherapy is not yet approved for this indication. This study presents the chemo-immunotherapy experience in patients with relapsed/progressive high-risk neuroblastoma treated within the off-label use program of the Neuroblastoma Committee of the French Society of Pediatric Oncology (SFCE). METHODS Dinutuximab beta (dB) was administered alongside temozolomide-topotecan (TOTEM) or temozolomide-irinotecan (TEMIRI) at first disease relapse/progression or topotecan-cyclophosphamide (TopoCyclo) at further relapse/progression. Real-world data on demographics, treatment, antitumor activity and safety was collected from all patients after inclusion in SACHA-France (NCT04477681), a prospective national registry, which documents safety and efficacy data on innovative anticancer therapies prescribed to patients ≤ 25 years old as compassionate or off-label use. RESULTS Between February 2021 and July 2023, 39 patients with confirmed relapsed/progressive high-risk neuroblastoma (median age 6 years, range 1-24) were treated with dB+TopoCyclo (n = 24) or dB+TOTEM/TEMIRI (n = 15) across 17 centers. In total, 163 chemo-immunotherapy cycles were administered, main toxicities were mild or moderate, with higher incidence of hematological adverse drug reactions with dB+TopoCyclo than dB+TOTEM/TEMIRI. Objective response rate was 42% for dB+TopoCyclo (CI95% 22-63%) and 40% for dB+TOTEM/TEMIRI (CI95% 16-68%). CONCLUSION Similar objective response rates for dB+TopoCyclo and dB+TOTEM/TEMIRI in patients with relapsed/progressive high-risk neuroblastoma emphasize the importance of chemo-immunotherapy, irrespective of the chemotherapy backbone.
Collapse
Affiliation(s)
- Patricia Raiser
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Gudrun Schleiermacher
- RTOP (Recherche Translationelle en Oncologie Pediatrique), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France; SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France
| | - Marion Gambart
- Department of Pediatric Oncology, Toulouse University Hospital, 31300 Toulouse, France
| | - Benoit Dumont
- Department of Pediatric Oncology, Institut d'Hématologie et d'Oncologie Pédiatrique/Centre Léon Bérard, 69008 Lyon, France
| | | | - Estelle Thebaud
- Pediatric Immuno-Hemato-Oncology Unit, CHU Nantes, 44000 Nantes, France
| | - Julie Tandonnet
- Pediatric Hematology-Oncology Department, Centre Hospitalier Universitaire (CHU), 33000 Bordeaux, France
| | - Claudia Pasqualini
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Stéphanie Proust
- Department of Pediatric Oncology, University Hospital, 49100 Angers, France
| | - Natacha Entz-Werle
- Pediatric Onco-Hematology Unit, University Hospital of Strasbourg, 67200 Strasbourg, France
| | - Isabelle Aerts
- RTOP (Recherche Translationelle en Oncologie Pediatrique), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France; SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), U830 INSERM, Institut Curie, PSL Research University, 75005 Paris, France
| | - Lee A Ndounga-Diakou
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Arnaud Petit
- Department of Pediatric Hematology and Oncology, Hôpital Armand Trousseau, 75012 Paris, France
| | - Chloe Puiseux
- Department of Pediatric Hemato-Oncology, University Hospital of Rennes, 35200 Rennes, France
| | - Camille Khanfar
- Department of Pediatric Oncology, CHU Amiens Picardie, 80054 Amiens, France
| | - Jeremie Rouger
- Department of Pediatric Hematology and Oncology, University Hospital of Caen, 14000 Caen, France
| | - Ludovic Mansuy
- Department of Pediatric Hematology-Oncology, Children's Hospital of Brabois, 54500 Vandoeuvre Les Nancy, France
| | - Joy Benadiba
- Department of Hemato-Oncology Pediatric, Nice University Hospital, 06000 Nice, France
| | - Frédéric Millot
- Department of Paediatric Haematology and Oncology, Centre Hospitalo-Universitaire de Poitiers, 86000 Poitiers, France
| | - Claire Pluchart
- Department of Paediatric Haematology and Oncology, Centre Hospitalo-Universitaire de Reims, 51100 Reims, France
| | - Salim Laghouati
- Pharmacovigilance Unit, Clinical Research Direction, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France; INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Gilles Vassal
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Dominique Valteau-Couanet
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Pablo Berlanga
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France.
| |
Collapse
|
44
|
Gorostegui M, Muñoz JP, Perez-Jaume S, Simao-Rafael M, Larrosa C, Garraus M, Salvador N, Lavarino C, Krauel L, Mañe S, Castañeda A, Mora J. Management of High-Risk Neuroblastoma with Soft-Tissue-Only Disease in the Era of Anti-GD2 Immunotherapy. Cancers (Basel) 2024; 16:1735. [PMID: 38730688 PMCID: PMC11083939 DOI: 10.3390/cancers16091735] [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: 04/09/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Neuroblastoma presents with two patterns of disease: locoregional or systemic. The poor prognostic risk factors of locoregional neuroblastoma (LR-NB) include age, MYCN or MDM2-CDK4 amplification, 11q, histology, diploidy with ALK or TERT mutations, and ATRX aberrations. Anti-GD2 immunotherapy has significantly improved the outcome of high-risk (HR) NB and is mostly effective against osteomedullary minimal residual disease (MRD), but less so against soft tissue disease. The question is whether adding anti-GD2 monoclonal antibodies (mAbs) benefits patients with HR-NB compounded by only soft tissue. We reviewed 31 patients treated at SJD for HR-NB with no osteomedullary involvement at diagnosis. All tumors had molecular genetic features of HR-NB. The outcome after first-line treatment showed 25 (80.6%) patients achieving CR. Thirteen patients remain in continued CR, median follow-up 3.9 years. We analyzed whether adding anti-GD2 immunotherapy to first-line treatment had any prognostic significance. The EFS analysis using Cox models showed a HR of 0.20, p = 0.0054, and an 80% decrease in the risk of relapse in patients treated with anti-GD2 immunotherapy in the first line. Neither EFS nor OS were significantly different by CR status after first-line treatment. In conclusion, adding treatment with anti-GD2 mAbs at the stage of MRD helps prevent relapse that unequivocally portends poor survival.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (M.G.); (J.P.M.); (M.S.-R.); (C.L.); (M.G.); (N.S.); (C.L.); (L.K.); (S.M.); (A.C.)
| |
Collapse
|
45
|
Mora J, Modak S, Kinsey J, Ragsdale CE, Lazarus HM. GM-CSF, G-CSF or no cytokine therapy with anti-GD2 immunotherapy for high-risk neuroblastoma. Int J Cancer 2024; 154:1340-1364. [PMID: 38108214 DOI: 10.1002/ijc.34815] [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: 06/14/2023] [Revised: 10/17/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
Colony-stimulating factors have been shown to improve anti-disialoganglioside 2 (anti-GD2) monoclonal antibody response in high-risk neuroblastoma by enhancing antibody-dependent cell-mediated cytotoxicity (ADCC). A substantial amount of research has focused on recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF) as an adjuvant to anti-GD2 monoclonal antibodies. There may be a disparity in care among patients as access to GM-CSF therapy and anti-GD2 monoclonal antibodies is not uniform. Only select countries have approved these agents for use, and even with regulatory approvals, access to these agents can be complex and cost prohibitive. This comprehensive review summarizes clinical data regarding efficacy and safety of GM-CSF, recombinant human granulocyte colony-stimulating factor (G-CSF) or no cytokine in combination with anti-GD2 monoclonal antibodies (ie, dinutuximab, dinutuximab beta or naxitamab) for immunotherapy of patients with high-risk neuroblastoma. A substantial body of clinical data support the immunotherapy combination of anti-GD2 monoclonal antibodies and GM-CSF. In contrast, clinical data supporting the use of G-CSF are limited. No formal comparison between GM-CSF, G-CSF and no cytokine has been identified. The treatment of high-risk neuroblastoma with anti-GD2 therapy plus GM-CSF is well established. Suboptimal efficacy outcomes with G-CSF raise concerns about its suitability as an alternative to GM-CSF as an adjuvant in immunotherapy for patients with high-risk neuroblastoma. While programs exist to facilitate obtaining GM-CSF and anti-GD2 monoclonal antibodies in regions where they are not commercially available, continued work is needed to ensure equitable therapeutic options are available globally.
Collapse
Affiliation(s)
- Jaume Mora
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Shakeel Modak
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joyce Kinsey
- Partner Therapeutics, Inc, Lexington, Massachusetts, USA
| | | | | |
Collapse
|
46
|
Fawzy M, Ahmed G, Youssef Y, Elkinaai N, Refaat A, Elahmadawy MA, Said F, Elmenawi S. Bilateral adrenal neuroblastoma: peculiar pattern of a rare pediatric presentation. Discov Oncol 2024; 15:115. [PMID: 38607453 PMCID: PMC11014822 DOI: 10.1007/s12672-024-00966-6] [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: 11/06/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Bilateral suprarenal neuroblastoma (BSN) is a rare presentation. Few previously published literature showed BSN patients to have favorable pattern and prognosis. This study aim was to evaluate clinical and biological features in relation to outcome of Egyptian patients with BSN. METHODS Included patients were diagnosed from 2007 to 2017, retrospectively. Tissue biopsy, imaging and bone marrow were evaluated at presentation. Clinical, demographic, biological variables and risk group were determined and analyzed in relation to overall (OS) and event-free-survival (EFS). RESULTS BSN patients (n = 33) represented 2% of hospital patients with neuroblastoma during the 10-year study period, 17 were males and 16 were females. Twenty-four patients (72.7%) were infants, and 9 patients (27.3%) were above 1 year of age (range: 1 month to 3 years). Metachronous disease was present in only one patient. Amplified MYCN was found in 10 patients. Initially, most patients (n = 25) had distant metastasis, 6 had stage 3 versus 2 stage 2. Fifteen were high risk (HR), 15 intermediate (IR), 1 low risk (LR) and 2 were undetermined due to inadequate tissue biopsy. Three-year OS for HR and IR patients were 40.5% and 83.9% versus 23.2% and 56.6% EFS; respectively. CONCLUSION BSN treatment is similar to unilateral disease. A more conservative surgical approach with adrenal tissue preservation on less extensive side should be considered. Biological variables and extent of disease are amongst the most important prognostic determinants. Future studies are warranted to further address the biologic profiling of BSN and highlight prognostic significance of size difference between both adrenal sides.
Collapse
Affiliation(s)
- Mohamed Fawzy
- Pediatric Oncology Department, Children's Cancer Hospital Egypt-57357, Cairo, Egypt
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Gehad Ahmed
- Faculty of Medicine, Department of General Surgery, Helwan University, Cairo, Egypt
- Surgical Oncology Department, Children's Cancer Hospital Egypt-57357, Cairo, Egypt
| | - Yasser Youssef
- Pediatric Oncology Department, Children's Cancer Hospital Egypt-57357, Cairo, Egypt
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Naglaa Elkinaai
- National Cancer Institute, Cairo University, Cairo, Egypt
- Pathology Department, Children's Cancer Hospital Egypt-57357, Cairo, Egypt
| | - Amal Refaat
- National Cancer Institute, Cairo University, Cairo, Egypt
- Radiodiagnosis Department, Children's Cancer Hospital Egypt-57357, Cairo, Egypt
| | - Mai Amr Elahmadawy
- National Cancer Institute, Cairo University, Cairo, Egypt
- Nuclear Medicine Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Fadwa Said
- Department of Clinical Pathology, Cairo University, Cairo, Egypt
| | - Salma Elmenawi
- Clinical Research Department, Children's Cancer Hospital Egypt-57357, 1 Seket Al-Emam Street, El-Madbah El-Kadeem Yard, El-Saida Zenab, Cairo, Egypt.
| |
Collapse
|
47
|
Moreno L, Weston R, Owens C, Valteau-Couanet D, Gambart M, Castel V, Zwaan CM, Nysom K, Gerber N, Castellano A, Laureys G, Ladenstein R, Rössler J, Makin G, Murphy D, Morland B, Vaidya S, Thebaud E, van Eijkelenburg N, Tweddle DA, Barone G, Tandonnet J, Corradini N, Chastagner P, Paillard C, Bautista FJ, Gallego Melcon S, De Wilde B, Marshall L, Gray J, Burchill SA, Schleiermacher G, Chesler L, Peet A, Leach MO, McHugh K, Hayes R, Jerome N, Caron H, Laidler J, Fenwick N, Holt G, Moroz V, Kearns P, Gates S, Pearson ADJ, Wheatley K. Bevacizumab, Irinotecan, or Topotecan Added to Temozolomide for Children With Relapsed and Refractory Neuroblastoma: Results of the ITCC-SIOPEN BEACON-Neuroblastoma Trial. J Clin Oncol 2024; 42:1135-1145. [PMID: 38190578 PMCID: PMC11003502 DOI: 10.1200/jco.23.00458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/25/2023] [Accepted: 10/05/2023] [Indexed: 01/10/2024] Open
Abstract
PURPOSE Outcomes for children with relapsed and refractory high-risk neuroblastoma (RR-HRNB) remain dismal. The BEACON Neuroblastoma trial (EudraCT 2012-000072-42) evaluated three backbone chemotherapy regimens and the addition of the antiangiogenic agent bevacizumab (B). MATERIALS AND METHODS Patients age 1-21 years with RR-HRNB with adequate organ function and performance status were randomly assigned in a 3 × 2 factorial design to temozolomide (T), irinotecan-temozolomide (IT), or topotecan-temozolomide (TTo) with or without B. The primary end point was best overall response (complete or partial) rate (ORR) during the first six courses, by RECIST or International Neuroblastoma Response Criteria for patients with measurable or evaluable disease, respectively. Safety, progression-free survival (PFS), and overall survival (OS) time were secondary end points. RESULTS One hundred sixty patients with RR-HRNB were included. For B random assignment (n = 160), the ORR was 26% (95% CI, 17 to 37) with B and 18% (95% CI, 10 to 28) without B (risk ratio [RR], 1.52 [95% CI, 0.83 to 2.77]; P = .17). Adjusted hazard ratio for PFS and OS were 0.89 (95% CI, 0.63 to 1.27) and 1.01 (95% CI, 0.70 to 1.45), respectively. For irinotecan ([I]; n = 121) and topotecan (n = 60) random assignments, RRs for ORR were 0.94 and 1.22, respectively. A potential interaction between I and B was identified. For patients in the bevacizumab-irinotecan-temozolomide (BIT) arm, the ORR was 23% (95% CI, 10 to 42), and the 1-year PFS estimate was 0.67 (95% CI, 0.47 to 0.80). CONCLUSION The addition of B met protocol-defined success criteria for ORR and appeared to improve PFS. Within this phase II trial, BIT showed signals of antitumor activity with acceptable tolerability. Future trials will confirm these results in the chemoimmunotherapy era.
Collapse
Affiliation(s)
- Lucas Moreno
- Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | - Guy Makin
- Central Manchester and Manchester Children's University Hospitals NHS Trust, Manchester, United Kingdom
| | - Dermot Murphy
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Bruce Morland
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - Sucheta Vaidya
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | | | | | - Deborah A Tweddle
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | | | | | | | | | | | | | | | | | - Lynley Marshall
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | - Juliet Gray
- University Hospital Southampton, Southampton, United Kingdom
| | | | | | - Louis Chesler
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | - Andrew Peet
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - Martin O Leach
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | - Kieran McHugh
- Great Ormond Street Hospital, London, United Kingdom
| | | | - Neil Jerome
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | | | | | | | - Grace Holt
- University of Birmingham, Birmingham, United Kingdom
| | | | - Pamela Kearns
- University of Birmingham, Birmingham, United Kingdom
| | - Simon Gates
- University of Birmingham, Birmingham, United Kingdom
| | - Andrew D J Pearson
- The Royal Marsden NHS Foundation Trust & Institute for Cancer Research, London, United Kingdom
| | | |
Collapse
|
48
|
Yankelevich M, Thakur A, Modak S, Chu R, Taub J, Martin A, Schalk D, Schienshang A, Whitaker S, Rea K, Lee DW, Liu Q, Shields AF, Cheung NKV, Lum LG. Targeting refractory/recurrent neuroblastoma and osteosarcoma with anti-CD3×anti-GD2 bispecific antibody armed T cells. J Immunother Cancer 2024; 12:e008744. [PMID: 38519053 PMCID: PMC10961524 DOI: 10.1136/jitc-2023-008744] [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] [Accepted: 02/28/2024] [Indexed: 03/24/2024] Open
Abstract
BACKGROUND The survival benefit observed in children with neuroblastoma (NB) and minimal residual disease who received treatment with anti-GD2 monoclonal antibodies prompted our investigation into the safety and potential clinical benefits of anti-CD3×anti-GD2 bispecific antibody (GD2Bi) armed T cells (GD2BATs). Preclinical studies demonstrated the high cytotoxicity of GD2BATs against GD2+cell lines, leading to the initiation of a phase I/II study in recurrent/refractory patients. METHODS The 3+3 dose escalation phase I study (NCT02173093) encompassed nine evaluable patients with NB (n=5), osteosarcoma (n=3), and desmoplastic small round cell tumors (n=1). Patients received twice-weekly infusions of GD2BATs at 40, 80, or 160×106 GD2BATs/kg/infusion complemented by daily interleukin-2 (300,000 IU/m2) and twice-weekly granulocyte macrophage colony-stimulating factor (250 µg/m2). The phase II segment focused on patients with NB at the dose 3 level of 160×106 GD2BATs/kg/infusion. RESULTS Of the 12 patients enrolled, 9 completed therapy in phase I with no dose-limiting toxicities. Mild and manageable cytokine release syndrome occurred in all patients, presenting as grade 2-3 fevers/chills, headaches, and occasional hypotension up to 72 hours after GD2BAT infusions. GD2-antibody-associated pain was minimal. Median overall survival (OS) for phase I and the limited phase II was 18.0 and 31.2 months, respectively, with a combined OS of 21.1 months. A phase I NB patient had a complete bone marrow response with overall stable disease. In phase II, 10 of 12 patients were evaluable: 1 achieved partial response, and 3 showed clinical benefit with prolonged stable disease. Over 50% of evaluable patients exhibited augmented immune responses to GD2+targets post-GD2BATs, as indicated by interferon-gamma (IFN-γ) EliSpots, Th1 cytokines, and/or chemokines. CONCLUSIONS This study demonstrated the safety of GD2BATs up to 160×106 cells/kg/infusion. Coupled with evidence of post-treatment endogenous immune responses, our findings support further investigation of GD2BATs in larger phase II clinical trials.
Collapse
Affiliation(s)
- Maxim Yankelevich
- St. Christopher's Hospital for Children, Philadelphia, Pennsylvania, USA
- Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Archana Thakur
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Roland Chu
- Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Jeffrey Taub
- Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Alissa Martin
- Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Dana Schalk
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Amy Schienshang
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Sarah Whitaker
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Katie Rea
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Daniel W Lee
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Qin Liu
- Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lawrence G Lum
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| |
Collapse
|
49
|
Gatz SA, Harttrampf AC, Brard C, Bautista F, André N, Abbou S, Rubino J, Rondof W, Deloger M, Rübsam M, Marshall LV, Hübschmann D, Nebchi S, Aerts I, Thebaud E, De Carli E, Defachelles AS, Paoletti X, Godin R, Miah K, Mortimer PGS, Vassal G, Geoerger B. Phase I/II Study of the WEE1 Inhibitor Adavosertib (AZD1775) in Combination with Carboplatin in Children with Advanced Malignancies: Arm C of the AcSé-ESMART Trial. Clin Cancer Res 2024; 30:741-753. [PMID: 38051741 DOI: 10.1158/1078-0432.ccr-23-2959] [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: 09/27/2023] [Revised: 11/06/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023]
Abstract
PURPOSE AcSé-ESMART Arm C aimed to define the recommended dose and activity of the WEE1 inhibitor adavosertib in combination with carboplatin in children and young adults with molecularly enriched recurrent/refractory malignancies. PATIENTS AND METHODS Adavosertib was administered orally, twice every day on Days 1 to 3 and carboplatin intravenously on Day 1 of a 21-day cycle, starting at 100 mg/m2/dose and AUC 5, respectively. Patients were enriched for molecular alterations in cell cycle and/or homologous recombination (HR). RESULTS Twenty patients (median age: 14.0 years; range: 3.4-23.5) were included; 18 received 69 treatment cycles. Dose-limiting toxicities were prolonged grade 4 neutropenia and grade 3/4 thrombocytopenia requiring transfusions, leading to two de-escalations to adavosertib 75 mg/m2/dose and carboplatin AUC 4; no recommended phase II dose was defined. Main treatment-related toxicities were hematologic and gastrointestinal. Adavosertib exposure in children was equivalent to that in adults; both doses achieved the cell kill target. Overall response rate was 11% (95% confidence interval, 0.0-25.6) with partial responses in 2 patients with neuroblastoma. One patient with medulloblastoma experienced unconfirmed partial response and 5 patients had stable disease beyond four cycles. Seven of these eight patients with clinical benefit had alterations in HR, replication stress, and/or RAS pathway genes with or without TP53 alterations, whereas TP53 pathway alterations alone (8/10) or no relevant alterations (2/10) were present in the 10 patients without benefit. CONCLUSIONS Adavosertib-carboplatin combination exhibited significant hematologic toxicity. Activity signals and identified potential biomarkers suggest further studies with less hematotoxic DNA-damaging therapy in molecularly enriched pediatric cancers.
Collapse
Affiliation(s)
- Susanne A Gatz
- Institute of Cancer and Genomic Sciences, University of Birmingham; Women's and Children's NHS Foundation Trust, Birmingham, United Kingdom
| | - Anne C Harttrampf
- Gustave Roussy Cancer Campus, Department of Pediatric and Adolescent Oncology, Villejuif, France
- Gustave Roussy Cancer Campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Caroline Brard
- Gustave Roussy Cancer Campus, Biostatistics and Epidemiology Unit, INSERM U1018, CESP, Université Paris-Saclay, Université Paris-Sud, UVSQ, Villejuif, France
| | - Francisco Bautista
- Hospital Niño Jesús, Department of Pediatric Oncology, Hematology and Stem Cell Transplantation, Madrid, Spain
| | - Nicolas André
- Hôpital de la Timone, AP-HM, Department of Pediatric Oncology, Marseille, France
- UMR INSERM 1068, CNRS UMR 7258, Aix Marseille Université U105, Marseille, Cancer Research Center (CRCM), Marseille, France
| | - Samuel Abbou
- Gustave Roussy Cancer Campus, Department of Pediatric and Adolescent Oncology, Villejuif, France
| | - Jonathan Rubino
- Gustave Roussy Cancer Campus, Clinical Research Direction, Villejuif, France
| | - Windy Rondof
- Gustave Roussy Cancer Campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
- Gustave Roussy Cancer Campus, Bioinformatics platform, Université Paris-Saclay, Villejuif, France
| | - Marc Deloger
- Gustave Roussy Cancer Campus, Bioinformatics platform, Université Paris-Saclay, Villejuif, France
| | - Marc Rübsam
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center
| | - Lynley V Marshall
- Royal Marsden Hospital & The Institute of Cancer Research, Paediatric and Adolescent Oncology Drug Development Unit, London, United Kingdom
| | - Daniel Hübschmann
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center
- Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem cell Technology and Experimental Medicine (HI-STEM); German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Souad Nebchi
- Gustave Roussy Cancer Campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Isabelle Aerts
- Institut Curie, SIREDO Oncology Center (Care, Innovation and research for children and AYA with cancer), PSL Research University, Paris, France
| | - Estelle Thebaud
- Centre Hospitalier Universitaire, Department of Pediatric Oncology, Nantes, France
| | - Emilie De Carli
- Centre Hospitalier Universitaire, Department of Pediatric Oncology, Angers, France
| | | | - Xavier Paoletti
- Gustave Roussy Cancer Campus, Biostatistics and Epidemiology Unit, INSERM U1018, CESP, Université Paris-Saclay, Université Paris-Sud, UVSQ, Villejuif, France
| | - Robert Godin
- AstraZeneca Oncology External R&D, Waltham, Massachusetts
| | - Kowser Miah
- Clinical Pharmacology and Quantitative Pharmacology, AstraZeneca, Waltham, Massachusetts
| | | | - Gilles Vassal
- Gustave Roussy Cancer Campus, Clinical Research Direction, Villejuif, France
| | - Birgit Geoerger
- Gustave Roussy Cancer Campus, Department of Pediatric and Adolescent Oncology, Villejuif, France
- Gustave Roussy Cancer Campus, INSERM U1015, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
50
|
Fenwick N, Weston R, Wheatley K, Hodgson J, Marshall L, Elliott M, Makin G, Ng A, Brennan B, Lowis S, Adamski J, Kilday JP, Cox R, Gattens M, Moore A, Trahair T, Ronghe M, Campbell M, Campbell H, Williams MW, Kirby M, Van Eijkelenburg N, Keely J, Scarpa U, Stavrou V, Fultang L, Booth S, Cheng P, De Santo C, Mussai F. PARC: a phase I/II study evaluating the safety and activity of pegylated recombinant human arginase BCT-100 in relapsed/refractory cancers of children and young adults. Front Oncol 2024; 14:1296576. [PMID: 38357205 PMCID: PMC10864630 DOI: 10.3389/fonc.2024.1296576] [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: 09/18/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Background The survival for many children with relapsed/refractory cancers remains poor despite advances in therapies. Arginine metabolism plays a key role in the pathophysiology of a number of pediatric cancers. We report the first in child study of a recombinant human arginase, BCT-100, in children with relapsed/refractory hematological, solid or CNS cancers. Procedure PARC was a single arm, Phase I/II, international, open label study. BCT-100 was given intravenously over one hour at weekly intervals. The Phase I section utilized a modified 3 + 3 design where escalation/de-escalation was based on both the safety profile and the complete depletion of arginine (defined as adequate arginine depletion; AAD <8μM arginine in the blood after 4 doses of BCT-100). The Phase II section was designed to further evaluate the clinical activity of BCT-100 at the pediatric RP2D determined in the Phase I section, by recruitment of patients with pediatric cancers into 4 individual groups. A primary evaluation of response was conducted at eight weeks with patients continuing to receive treatment until disease progression or unacceptable toxicity. Results 49 children were recruited globally. The Phase I cohort of the trial established the Recommended Phase II Dose of 1600U/kg iv weekly in children, matching that of adults. BCT-100 was very well tolerated. No responses defined as a CR, CRi or PR were seen in any cohort within the defined 8 week primary evaluation period. However a number of these relapsed/refractory patients experienced prolonged radiological SD. Conclusion Arginine depletion is a clinically safe and achievable strategy in children with cancer. The RP2D of BCT-100 in children with relapsed/refractory cancers is established at 1600U/kg intravenously weekly and can lead to sustained disease stability in this hard to treat population. Clinical trial registration EudraCT, 2017-002762-44; ISRCTN, 21727048; and ClinicalTrials.gov, NCT03455140.
Collapse
Affiliation(s)
- Nicola Fenwick
- Children’s Cancer Trials Team, Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, United Kingdom
| | - Rebekah Weston
- Children’s Cancer Trials Team, Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, United Kingdom
| | - Keith Wheatley
- Children’s Cancer Trials Team, Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, United Kingdom
| | - Jodie Hodgson
- Children’s Cancer Trials Team, Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, United Kingdom
| | | | - Martin Elliott
- Leeds Teaching Hospital, St James University Hospital, Leeds, United Kingdom
| | - Guy Makin
- Royal Manchester Children’s Hospital, Manchester, United Kingdom
| | - Antony Ng
- Bristol Royal Hospital for Children, Bristol, United Kingdom
| | | | - Stephen Lowis
- Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Jenny Adamski
- Birmingham Children’s Hospital, Birmingham, United Kingdom
| | - John Paul Kilday
- Royal Manchester Children’s Hospital, Manchester, United Kingdom
| | - Rachel Cox
- Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Mike Gattens
- Addenbrookes Hospital, Cambridge, United Kingdom
| | - Andrew Moore
- Queensland Children’s Hospital, Brisbane, QLD, Australia
| | - Toby Trahair
- Sydney Children’s Hospital, Sydney, NSW, Australia
| | - Milind Ronghe
- Royal Hospital for Children, Glasgow, United Kingdom
| | | | - Helen Campbell
- Royal Manchester Children’s Hospital, Manchester, United Kingdom
| | | | - Maria Kirby
- Michael Rice Cancer Centre, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| | | | - Jennifer Keely
- Children’s Cancer Trials Team, Cancer Research UK Clinical Trials Unit (CRCTU), University of Birmingham, Birmingham, United Kingdom
| | - Ugo Scarpa
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Victoria Stavrou
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Livingstone Fultang
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Sarah Booth
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Paul Cheng
- Bio-Cancer Treatment International, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Francis Mussai
- Birmingham Children’s Hospital, Birmingham, United Kingdom
| |
Collapse
|