1
|
Knight TE, Broglie L, Sharma A, Qayed M, Yanik GA. Why are Higher CD34+ Cell Doses Associated with Improved Outcomes among Pediatric Patients Undergoing Autologous Hematopoietic Stem Cell Transplant for Central Nervous System Tumors - But Not for High-Risk Neuroblastoma? Pediatr Hematol Oncol 2025; 42:63-67. [PMID: 39501613 PMCID: PMC11745919 DOI: 10.1080/08880018.2024.2420924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 01/21/2025]
Affiliation(s)
- Tristan E. Knight
- Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA
- Division of Hematology and Oncology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA
| | - Larisa Broglie
- CIBMTR (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Emory University and Children’s Healthcare of Atlanta, Atlanta, GA
| | - Gregory A. Yanik
- Mott Children’s Hospital, University of Michigan Medical Center, Ann Arbor, MI
| |
Collapse
|
2
|
Swaminathan VV, Uppuluri R, Meena SK, Varla H, Chandar R, Raj R. Haploidentical stem cell transplantation in newly diagnosed high-risk neuroblastoma: Results from a single-arm, prospective study from India. Indian J Cancer 2025; 62:32-36. [PMID: 40377603 DOI: 10.4103/ijc.ijc_999_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 06/16/2022] [Indexed: 05/18/2025]
Abstract
BACKGROUND With increasing data on graft-versus-tumor (GVT) effect in neuroblastoma, we aimed to evaluate the possibility of haploidentical stem cell transplantation (HSCT) as a technique to induce GVT and thereby improve outcomes in high-risk neuroblastoma. PATIENTS AND METHODS We performed a prospective, single-arm study and included children from 18 months to 18 years of age diagnosed with high-risk neuroblastoma and who underwent a haploidentical HSCT from a parent donor. All children were started on induction chemotherapy as per the SIOP-Europa-Neuroblastoma (SIOPEN) protocol, followed by assessment and surgery when feasible. Conditioning in the unmanipulated graft included thiotepa/fludarabine/busulfan. With a T-depleted graft, conditioning included rabbit anti-thymocyte globulin/fludarabine/thiotepa/melphalan. RESULTS Of the five children, one child received T-depleted and four children received T-replete stem cells with posttransplant cyclophosphamide, with 100% engraftment, no regimen-related toxicities, and documented remission. Grade I/II skin GVHD occurred in all children who responded to steroids. We administered involved-field radiotherapy in four children around D + 60-75 post-HSCT, followed by 13-cis retinoic acid. At 1 year post-HSCT, three had isolated central nervous system relapse; one child had isolated pelvic bone relapse. One child relapsed 2 months post-HSCT. Progression-free survival was 12 months in 4/5 children and 6 months in one child. All except one child succumbed to progressive disease post relapse. CONCLUSION Children with high-risk neuroblastoma continued to relapse within a year post-HSCT in our cohort. Significant improvement in outcomes was not demonstrated with haploidentical HSCT. We urgently need new strategies including deeper remission induction with upfront metaiodobenzylguanidine (MIBG) therapy followed by HSCT to improve the outcomes.
Collapse
Affiliation(s)
- Venkateswaran V Swaminathan
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, Tamil Nadu, India
| | | | | | | | | | | |
Collapse
|
3
|
Y KN, Arjunan A, Maigandan D, Dharmarajan A, Perumalsamy LR. Advances and challenges in therapeutic resistant biomarkers of neuroblastoma: A comprehensive review. Biochim Biophys Acta Rev Cancer 2024; 1879:189222. [PMID: 39577750 DOI: 10.1016/j.bbcan.2024.189222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Therapeutic resistance is one of the significant challenges in Neuroblastoma. Owing to its molecular diversity, the therapeutic resistance mechanisms of Neuroblastoma are highly complicated. The traditional chemo and radio therapeutics fail to provide adequate solutions to the treatment resistance, demanding in-depth research to improvise the existing prognostic and therapeutic regimens. To address this knowledge gap, several investigations are being employed, such as unravelling the molecular signalling mechanisms involved in drug resistance at genomics and proteomics levels, development of biomarkers for assessing the therapeutic success, development of novel drug targets for cancer stem cells, targeted immunotherapy and combination therapies. This review collates the ongoing research efforts to address the challenges faced in Neuroblastoma treatment resistance and uncovers the importance of transitioning biomarker discoveries into clinical practice.
Collapse
Affiliation(s)
- Krithicaa Narayanaa Y
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Amrutha Arjunan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Devi Maigandan
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| | - Arun Dharmarajan
- Sri Ramachandra Faculty of Clinical Research, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India; Curtin Health Innovation Research Institute, Curtin University, Bentley, 6102 Perth, Western Australia, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia; School of Human Sciences, The University of Western Australia, Nedlands, Western Australia, Australia.
| | - Lakshmi R Perumalsamy
- Department of Biomedical Sciences, Sri Ramachandra Faculty of Biomedical Sciences & Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India.
| |
Collapse
|
4
|
Yan Z, Wu Y, Chen Y, Xu J, Zhang X, Yin Q. A clinical prediction model for distant metastases of pediatric neuroblastoma: an analysis based on the SEER database. Front Pediatr 2024; 12:1417818. [PMID: 39363969 PMCID: PMC11447546 DOI: 10.3389/fped.2024.1417818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
Background Patients with distant metastases from neuroblastoma (NB) usually have a poorer prognosis, and early diagnosis is essential to prevent distant metastases. The aim was to develop a machine-learning model for predicting the risk of distant metastasis in patients with neuroblastoma to aid clinical diagnosis and treatment decisions. Methods We built a predictive model using data from the Surveillance, Epidemiology, and End Results (SEER) database from 2010 to 2018 on 1,542 patients with neuroblastoma. Seven machine-learning methods were employed to forecast the likelihood of neuroblastoma distant metastases. Univariate and multivariate logistic regression analyses were used to identify independent risk factors for building machine learning models. Secondly, the subject operating characteristic area under the curve (AUC), Precision-Recall (PR) curves, decision curve analysis (DCA), and calibration curves were used to assess model performance. To further explain the optimal model, the Shapley summation interpretation method (SHAP) was applied. Ultimately, the best model was used to create an online calculator that estimates the likelihood of neuroblastoma distant metastases. Results The study included 1,542 patients with neuroblastoma, multifactorial logistic regression analysis showed that age, histology, tumor size, tumor grade, primary site, surgery, chemotherapy, and radiotherapy were independent risk factors for distant metastasis of neuroblastoma (P < 0.05). Logistic regression (LR) was found to be the optimal algorithm among the seven constructed, with the highest AUC values of 0.835 and 0.850 in the training and validation sets, respectively. Finally, we used the logistic regression model to build a network calculator for distant metastasis of neuroblastoma. Conclusion The study developed and validated a machine learning model based on clinical and pathological information for predicting the risk of distant metastasis in patients with neuroblastoma, which may help physicians make clinical decisions.
Collapse
Affiliation(s)
- Zhiwei Yan
- Department of Paediatric Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yumeng Wu
- Cancer Research Center Nantong, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Yuehua Chen
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jian Xu
- Department of Medical Oncology, Nantong Second Peoples Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiubing Zhang
- Department of Medical Oncology, Nantong Second Peoples Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Qiyou Yin
- Department of Pediatric Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
5
|
Pearson H, Bell C, Cox K, Kayum C, Knox L, Gibson F, Myall M, Darlington AS, Potter E, Bird N. Integration of patient and public involvement in a doctoral research study using the research cycle. RESEARCH INVOLVEMENT AND ENGAGEMENT 2024; 10:87. [PMID: 39123249 PMCID: PMC11316368 DOI: 10.1186/s40900-024-00620-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Patient and public involvement (PPI) in research is widely acknowledged as essential to achieving successful and impactful research. Despite this acknowledgement, there are limited reports on how to approach and apply meaningful PPI throughout the research cycle and how to address challenges for researchers such as doctoral students, particularly when undertaking research on sensitive topics. This paper provides insights and examples for researchers new to PPI, on the impact of active PPI and recommendations for building and developing a PPI group in a paediatric focused doctoral research study with bereaved parents and carers. METHODS PPI was informed by the research cycle. The GRIPP2 short-form checklist was used to report PPI. The research was funded by the National Institute for Health and Care Research. RESULTS PPI enhanced the research through input into the study design, recruitment, co-design of the study website and branding; and ethics amendments to increase participation in response to the COVID-19 pandemic. The literature review was extended to incorporate a PPI consultation phase and members contributed to data analysis. A flexible approach enabled involvement to develop iteratively throughout the research study, resulting in changes being made to enhance the study design and outcomes. CONCLUSION This paper contributes to the limited knowledge base on embedding PPI into a doctoral research study and within the paediatric setting specifically working in partnership with bereaved parents and carers. Employing an adaptive approach to meet individual PPI needs, building a trusting and respectful partnership, creating shared ownership and investment in the research, are essential components to successful PPI.
Collapse
Affiliation(s)
- Helen Pearson
- School of Health Sciences, University of Southampton, Southampton, UK.
- The Oak Centre for Children and Young People, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK.
| | - Carol Bell
- Member of the REDMAPP Patient Public Involvement Group, London, UK
| | - Karl Cox
- Member of the REDMAPP Patient Public Involvement Group, London, UK
| | - Catherine Kayum
- Member of the REDMAPP Patient Public Involvement Group, London, UK
| | - Leona Knox
- Member of the REDMAPP Patient Public Involvement Group, London, UK
- Solving Kids' Cancer UK, London, UK
| | - Faith Gibson
- Centre for Outcomes and Experience Research in Children's Health, Illness and Disability (ORCHID), Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- School of Health Sciences, University of Surrey, Guildford, Surrey, UK
| | - Michelle Myall
- School of Health Sciences, University of Southampton, Southampton, UK
| | | | - Emma Potter
- The Oak Centre for Children and Young People, The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | - Nicholas Bird
- Member of the REDMAPP Patient Public Involvement Group, London, UK
- Solving Kids' Cancer UK, London, UK
| |
Collapse
|
6
|
Chen F, He B, Wang Y. Bone metastases among newly diagnosed cancer patients: a population-based study. Postgrad Med J 2024; 100:569-577. [PMID: 38497290 DOI: 10.1093/postmj/qgae036] [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: 08/25/2023] [Revised: 01/18/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024]
Abstract
PURPOSE (i) To analyze age-adjusted incidence rates of synchronous bone metastases diagnosed alongside primary malignancy from 2010 to 2018 in the US population, (ii) determine the incidence proportions (IPs) and characteristics of synchronous bone metastases among newly diagnosed cancer patients in the USA especially pediatric cases, and (iii) assess the implications of synchronous bone metastases on cancer patient's survival, and identify the survival risk factors for these cancer patients. METHODS Utilizing data from the Surveillance, Epidemiology, and End Results (SEER) program (2010-2018), we calculated age-adjusted IPs and annual percentage change (APC), and employed logistic regression and Cox regression models for our analysis. RESULTS 3 300 736 cancer patients were identified. The age-adjusted incidence rates of synchronous bone metastases increased from 2010 (18.04/100 000) to 2018 (20.89/100 000; APC: 2.3, 95% confidence interval [CI], 1.4-3.1), but decreased in lung cancer (average APC: -1.0, 95% CI, -1.8 to -0.3). The highest IPs were observed in pediatric neuroblastoma (43.2%; 95% CI, 39.8%-46.7%) and adult small cell carcinoma (23.1%; 95% CI, 22.7%-23.4%). Multivariate logistic analyses revealed that primary tumor characteristics were correlated with higher bone metastases risk. Survival analyses also showed varied prognostic outcomes based on metastasis sites and demographics among cancer patients. Landmark analyses further indicated among long-term cancer survivors (≥3 and ≥5 years), patients with de novo bone metastases had the poorest survival rates compared with those with other synchronous metastases (P < 0.001). CONCLUSION This study provides a population-based estimation of the incidence and prognosis for synchronous bone metastases. Our findings highlight the need for early identification of high-risk groups and multidisciplinary approaches to improve prognosis of cancer patients with de novo bone metastases.
Collapse
Affiliation(s)
- Feng Chen
- Department of Orthopedics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Bo He
- Department of Orthopedics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yang Wang
- Department of Orthopedics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| |
Collapse
|
7
|
Djos A, Svensson J, Gaarder J, Umapathy G, Nilsson S, Ek T, Vogt H, Georgantzi K, Öra I, Träger C, Kogner P, Martinsson T, Fransson S. Loss of Chromosome Y in Neuroblastoma Is Associated With High-Risk Disease, 11q-Deletion, and Telomere Maintenance. Genes Chromosomes Cancer 2024; 63:e23260. [PMID: 39031441 DOI: 10.1002/gcc.23260] [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: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024] Open
Abstract
Neuroblastoma (NB) is a heterogeneous childhood cancer with a slightly higher incidence in boys than girls, with the reason for this gender disparity unknown. Given the growing evidence for the involvement of loss of the Y chromosome (LoY) in male diseases including cancer, we investigated Y chromosome status in NB. Male NB tumor samples from a Swedish cohort, analyzed using Cytoscan HD SNP-microarray, were selected. Seventy NB tumors were analyzed for aneuploidy of the Y chromosome, and these data were correlated with other genetic, biological, and clinical parameters. LoY was found in 21% of the male NB tumors and it was almost exclusively found in those with high-risk genomic profiles. Furthermore, LoY was associated with increased age at diagnosis and enriched in tumors with 11q-deletion and activated telomere maintenance mechanisms. In contrast, tumors with an MYCN-amplified genomic profile retained their Y chromosome. The understanding of LoY in cancer is limited, making it difficult to conclude whether LoY is a driving event in NB or function of increased genomic instability. Gene expression analysis of Y chromosome genes in male NB tumors showed low expression of certain genes correlating with worse overall survival. KDM5D, encoding a histone demethylase stands out as an interesting candidate for further studies. LoY has been shown to impact the epigenomic layer of autosomal loci in nonreproductive tissues, and KDM5D has been reported as downregulated and/or associated with poor survival in different malignancies. Further studies are needed to explore the mechanisms and functional consequences of LoY in NB.
Collapse
Affiliation(s)
- Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johanna Svensson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jennie Gaarder
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ganesh Umapathy
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Torben Ek
- Children's Cancer Centre, Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hartmut Vogt
- Crown Princess Victoria Children's Hospital, Division of Children's and Women's Health, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Kleopatra Georgantzi
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Ingrid Öra
- Department of Pediatric Oncology, Skåne University Hospital, Lund, Sweden
| | - Catarina Träger
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, and Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Feng L, Zhou Z, Liu J, Yao S, Wang C, Zhang H, Xiong P, Wang W, Yang J. 18F-FDG PET/CT-Based Radiomics Nomogram for Prediction of Bone Marrow Involvement in Pediatric Neuroblastoma: A Two-Center Study. Acad Radiol 2024; 31:1111-1121. [PMID: 37643929 DOI: 10.1016/j.acra.2023.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023]
Abstract
RATIONALE AND OBJECTIVES To assess the predictive ability of an 18F-FDG PET/CT-based radiomics nomogram for bone marrow involvement in pediatric neuroblastoma. MATERIALS AND METHODS A total of 241 neuroblastoma patients who underwent 18F-FDG PET/CT at two medical centers were retrospectively evaluated. Data from center A (n = 200) were randomized into a training cohort (n = 140) and an internal validation cohort (n = 60), while data from center B (n = 41) constituted the external validation cohort. For each patient, two regions of interest were defined using the tumor and axial skeleton. The clinical factors and radiomics features were derived to construct the clinical and radiomics models. The radiomics nomogram was built by combining clinical factors and radiomics features. The area under the receiver operating characteristic curves (AUCs) were used to assess the performance of the models. RESULTS Radiomics models created from tumor and axial skeleton achieved AUCs of 0.773 and 0.900, and the clinical model had an AUC of 0.858 in the training cohort. By incorporating clinical risk factors and axial skeleton-based radiomics features, the AUC of the radiomics nomogram in the training cohort, internal validation cohort, and external validation cohort was 0.932, 0.887, and 0.733, respectively. CONCLUSION The axial skeleton-based radiomics model performed better than the tumor-based radiomics model in predicting bone marrow involvement. Moreover, the radiomics nomogram showed that combining axial skeleton-based radiomics features with clinical risk factors improved their performance.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Ziang Zhou
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Jun Liu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Shuang Yao
- Department of Nuclear Medicine, Beijing Fengtai YouAnMen Hospital, Beijing, China (S.Y.)
| | - Chao Wang
- Department of Clinical Research, SinoUnion Healthcare Inc., Beijing, China (C.W.)
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China (H.Z.)
| | - Pingxiang Xiong
- Nanchang Rimag Medical Diagnosis Center, Nanchang, China (P.X.)
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.)
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing 100050, China (L.F., Z.Z., J.L., W.W., J.Y.).
| |
Collapse
|
9
|
Wang H, He L, Chen X, Ding S, Xie M, Cai J. Predicting Bone Marrow Metastasis in Neuroblastoma: An Explainable Machine Learning Approach Using Contrast-Enhanced Computed Tomography Radiomics Features. Technol Cancer Res Treat 2024; 23:15330338241290386. [PMID: 39440370 PMCID: PMC11500234 DOI: 10.1177/15330338241290386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
PURPOSE To predict bone marrow metastasis in neuroblastoma using contrast-enhanced computed tomography (CECT) radiomics features and explainable machine learning. METHODS This cohort study retrospectively included a total of 345 neuroblastoma patients who underwent testing for bone marrow metastatic status. Tumor lesions on CECT images were delineated by two radiologists, and 1409 radiomics features were extracted. Correlation analysis, Least Absolute Shrinkage and Selection Operator regression, and one-way analysis of variance were used to identify radiomics features associated with bone marrow metastasis. A predictive model for bone marrow metastasis was then developed using the support vector machine algorithm based on the selected radiomics features. The performance of the radiomics model was evaluated using the area under the curve (AUC), 95% confidence interval (CI), accuracy, sensitivity, and specificity. RESULTS The radiomics model included 16 features, with a predominant focus on texture features (12/16, 75%). In the training set, the model demonstrated an AUC of 0.891 (95% CI: 0.848-0.933), an accuracy of 0.831 (95% CI: 0.829-0.832), a sensitivity of 0.893 (95% CI: 0.840-0.946), and a specificity of 0.757 (95% CI: 0.677-0.837). In the test set, the AUC, accuracy, sensitivity, and specificity were 0.807 (95% CI: 0.720-0.893), 0.767 (95% CI: 0.764-0.770), 0.696 (95% CI: 0.576-0.817), and 0.851 (95% CI: 0.749-0.953), respectively. CONCLUSION Radiomics features extracted from CECT images are associated with the presence of bone marrow metastasis in neuroblastoma, providing potential new imaging biomarkers for predicting bone marrow metastasis in this disease.
Collapse
Affiliation(s)
- Haoru Wang
- 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
| | - Ling He
- 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
| | - Xin Chen
- 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
| | - Shuang Ding
- 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
| | - Mingye Xie
- 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
- 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
|
10
|
Knight TE, Ahn KW, Hebert KM, Atshan R, Wall DA, Chiengthong K, Lund TC, Prestidge T, Rangarajan HG, Dvorak CC, Auletta JJ, Kent M, Hashem H, Talano JA, Rotz SJ, Fraint E, Myers KC, Leung W, Sharma A, Bhatt NS, Driscoll TA, Yu LC, Schultz KR, Qayed M, Broglie L, Eapen M, Yanik GA. No impact of CD34 + cell dose on outcome among children undergoing autologous hematopoietic stem cell transplant for high-risk neuroblastoma. Bone Marrow Transplant 2023; 58:1390-1393. [PMID: 37666957 PMCID: PMC11221560 DOI: 10.1038/s41409-023-02092-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023]
Affiliation(s)
- Tristan E Knight
- Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA, USA
- Division of Hematology and Oncology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Kwang Woo Ahn
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kyle M Hebert
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Rasha Atshan
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Donna A Wall
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Kanhatai Chiengthong
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Troy C Lund
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, University of Minnesota, Minneapolis, MN, USA
| | - Tim Prestidge
- Blood and Cancer Centre, Starship Children's Hospital, Auckland, New Zealand
| | - Hemalatha G Rangarajan
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplantation, Nationwide Children's Hospital, Columbus, OH, USA
| | - Christopher C Dvorak
- Division of Pediatric Allergy, Immunology & Bone Marrow Transplantation, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA
| | - Jeffery J Auletta
- CIBMTR® (Center for International Blood and Marrow Transplant Research), National Marrow Donor Program/Be The Match, Minneapolis, MN, USA
- Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital, Columbus, OH, USA
| | - Michael Kent
- Atrium Health Levine Children's, Charlotte, NC, USA
| | - Hasan Hashem
- Division of Pediatric Hematology/Oncology and Bone Marrow Transplantation, King Hussein Cancer Center, Amman, Jordan
| | - Julie-An Talano
- Department of Pediatric Hematology Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Seth J Rotz
- Department of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, OH, USA
| | - Ellen Fraint
- Division of Pediatric Hematology, Oncology, and Cellular Therapy, The Children's Hospital at Montefiore, Bronx, NY, USA
| | - Kasiani C Myers
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Wing Leung
- Duke-NUS Medical School, Singapore, Singapore
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Neel S Bhatt
- Division of Hematology and Oncology, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Timothy A Driscoll
- Duke University Pediatric Transplantation and Cellular Therapy, Durham, NC, USA
| | - Lolie C Yu
- Division of Hematology/Oncology & HSCT, The Center for Cancer and Blood Disorders, Children's Hospital, Louisiana State University Medical Center, New Orleans, LA, USA
| | - Kirk R Schultz
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, British Columbia's Children's Hospital, The University of British Columbia, Vancouver, BC, Canada
| | - Muna Qayed
- Aflac Cancer and Blood Disorders Center, Emory University and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Larisa Broglie
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Division of Pediatric Hematology/Oncology/Blood and Marrow Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Mary Eapen
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Gregory A Yanik
- Mott Children's Hospital, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Wang P, Li T, Liu Z, Jin M, Su Y, Zhang J, Jing H, Zhuang H, Li F. [ 18F]MFBG PET/CT outperforming [ 123I]MIBG SPECT/CT in the evaluation of neuroblastoma. Eur J Nucl Med Mol Imaging 2023; 50:3097-3106. [PMID: 37160439 DOI: 10.1007/s00259-023-06221-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/02/2023] [Indexed: 05/11/2023]
Abstract
PURPOSE Iodine 123 labeled meta-iodobenzylguanidine ([123I]MIBG) scan with SPECT/CT imaging is one of the most commonly used imaging modalities in the evaluation of neuroblastoma. [18F]-meta-fluorobenzylguanidine ([18F]MFBG) is a novel positron emission tomography (PET) tracer which was reported to have a similar biodistribution to [123I]MIBG. However, the experience of using [18F]MFBG PET/CT in the evaluation of patients with neuroblastoma is limited. This preliminary investigation aims to assess the efficacy of [18F]MFBG PET/CT in the evaluation of neuroblastomas in comparison to [123I]MIBG scans with SPECT/CT. MATERIALS AND METHODS In this prospective, single-center study, 40 participants (mean age 6.0 ± 3.7 years) with history of neuroblastoma were enrolled. All children underwent both [123I]MIBG SPECT/CT and [18F]MFBG PET/CT studies. The number of lesions and the Curie scores revealed by each imaging method were recorded. RESULTS Six patients had negative findings on both [123I]MIBG and [18F]MFBG studies. Four of the 34 patients (11.8%) were negative on [123I]MIBG but positive on [18F]MFBG, while 30 patients were positive on both [123I]MIBG and [18F]MFBG studies. In these 34 patients, [18F]MFBG PET/CT identified 784 lesions while [123I]MIBG SPECT/CT detected 532 lesions (p < 0.001). The Curie scores obtained from [18F]MFBG PET/CT (11.32 ± 8.18, range 1-27) were statistically higher (p < 0.001) than those from [123I]MIBG SPECT/CT (7.74 ± 7.52, range 0-26). 30 of 34 patients (88.2%) with active disease on imaging had higher Curie scores based on the [18F]MFBG study than on the [123I]MIBG imaging. CONCLUSION [18F]MFBG PET/CT shows higher lesion detection rate than [123I]MIBG SPECT/CT in the evaluation of pediatric patients with neuroblastoma. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov : NCT05069220 (Registered: 25 September 2021, retrospectively registered); Institute Review Board of Peking Union Medical College Hospital: ZS-2514.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China
| | - Tuo Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China
| | - Zhikai Liu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, People's Republic of China
| | - Mei Jin
- Department of Medical Oncology, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China
| | - Yan Su
- Department of Medical Oncology, Pediatric Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, People's Republic of China.
| | - Jingjing Zhang
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hongli Jing
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China.
| | - Hongming Zhuang
- Department of Radiology, Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Fang Li
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China.
| |
Collapse
|
12
|
Jahangiri L. Metastasis in Neuroblastoma and Its Link to Autophagy. Life (Basel) 2023; 13:life13030818. [PMID: 36983973 PMCID: PMC10056181 DOI: 10.3390/life13030818] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Neuroblastoma is a paediatric malignancy originating from the neural crest that commonly occurs in the abdomen and adrenal gland, leading to cancer-related deaths in children. Distant metastasis can be encountered at diagnosis in greater than half of these neuroblastoma patients. Autophagy, a self-degradative process, plays a key role in stress-related responses and the survival of cells and has been studied in neuroblastoma. Accordingly, in the early stages of metastasis, autophagy may suppress cancer cell invasion and migration, while its role may be reversed in later stages, and it may facilitate metastasis by enhancing cancer cell survival. To that end, a body of literature has revealed the mechanistic link between autophagy and metastasis in neuroblastoma in multiple steps of the metastatic cascade, including cancer cell invasion and migration, anoikis resistance, cancer cell dormancy, micrometastasis, and metastatic outbreak. This review aims to take a step forward and discuss the significance of multiple molecular players and compounds that may link autophagy to metastasis and map their function to various metastatic steps in neuroblastoma.
Collapse
Affiliation(s)
- Leila Jahangiri
- School of Science and Technology, Nottingham Trent University, Clifton Site, Nottingham NG11 8NS, UK
- Division of Cellular and Molecular Pathology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
13
|
Liu X, Wang Z, Xiong X, Li C, Wu Y, Su M, Yang S, Zeng M, Weng W, Huang K, Zhou D, Fang J, Xu L, Li P, Zhu Y, Qiu K, Ma Y, Lei J, Li Y. Arsenic Trioxide inhibits Activation of Hedgehog Pathway in Human Neuroblastoma Cell Line SK-N-BE(2) Independent of Itraconazole. Anticancer Agents Med Chem 2023; 23:2217-2224. [PMID: 37888819 DOI: 10.2174/0118715206259952230919173611] [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: 04/28/2023] [Revised: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Neuroblastoma (NB) remains associated with a low overall survival rate over the long term. Abnormal activation of the Hedgehog (HH) signaling pathway can activate the transcription of various downstream target genes that promote NB. Both arsenic trioxide (ATO) and itraconazole (ITRA) can inhibit tumor growth. OBJECTIVE To determine whether ATO combined with ITRA can be used to treat NB with HH pathway activation, we examined the effects of ATO and ITRA monotherapy or combined inhibition of the HH pathway in NB. METHODS Analysis of CCK8 and flow cytometry showed cell inhibition and cell cycle, respectively. Real-time PCR analysis was conducted to assess the mRNA expression of HH pathway. RESULTS We revealed that as concentrations of ATO and ITRA increased, the killing effects of both agents on SK-N-BE(2) cells became more apparent. During G2/M, the cell cycle was largely arrested by ATO alone and combined with ITRA, and in the G0/G1 phase by ITRA alone. In the HH pathway, ATO inhibited the transcription of the SHH, PTCH1, SMO and GLI2 genes, however, ITRA did not. Instead of showing synergistic effects in a combined mode, ITRA decreased ATO inhibitory effects. CONCLUSION We showed that ATO is an important inhibitor of HH pathway but ITRA can weaken the inhibitory effect of ATO. This study provides an experimental evidence for the clinical use of ATO and ITRA in the treatment of NB with HH pathway activation in cytology.
Collapse
Affiliation(s)
- Xiaoshan Liu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhixuan Wang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xilin Xiong
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chunmou Li
- Department of Pediatrics, the Seventh Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Yu Wu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Mingwei Su
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shu Yang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Meilin Zeng
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenjun Weng
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ke Huang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dunhua Zhou
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianpei Fang
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lvhong Xu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Peng Li
- South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yafeng Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Kunyin Qiu
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuhan Ma
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiaying Lei
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yang Li
- Pediatric Hematology/Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
14
|
Nanyanga I, Kwikiriza GK, Atwiine B, Namazzi R, Musiime V, Kambugu JB, van Heerden J. A retrospective evaluation of the presentation, prognostic factors and outcomes of neuroblastoma in Ugandan children. Pediatr Hematol Oncol 2022; 40:281-299. [PMID: 36562399 DOI: 10.1080/08880018.2022.2159593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor of childhood. The complete burden and outcomes in Uganda are unknown. The study was a multicenter retrospective chart review of children aged between 0 to 15 years diagnosed with NB from 2010 to 2020. Demographic, clinical and tumor-related characteristics were extracted for analysis. Kaplan-Meier survival curves and Cox regression models were used to determine the one-year overall survival (OS) and identify prognostic factors. Seventy-five patients were evaluated, with a median age at diagnosis of 48 months (IQR 26-108 months). Fever (74.7%), weight loss (74.7%), high blood pressure (70.3%) and abdominal swelling/mass (65.3%) were the most common features at diagnosis. Suprarenal tumors (52%) and stage 4 disease (70.7%) were also common. The one-year OS was 60.0% (95%CI 56.8%; 64.3%) with a median survival time of 12.6 months (95% CI: 8.1; 20.8). The one-year OS for non-metastatic and metastatic disease was 67.3% and 42.6% (p = 0.11) respectively. Leukocytosis (p < 0.001) at diagnosis was of prognostic significance while clinical remission after induction chemotherapy (p < 0.001) provided survival advantages. Children who received maintenance chemotherapy had a longer median survival time of 38.5 months (range 10.8-69.5). Age (p = 0.001), lung metastasis (p < 0.001), and leukocytosis (p < 0.001) remained significant on multivariate analysis. In this Ugandan study, leukocytosis was a clinical predictor of prognosis, metastatic disease had management challenges and maintenance chemotherapy prolonged the survival time but not OS.
Collapse
Affiliation(s)
- Irene Nanyanga
- Department of Pediatric Oncology, Uganda Cancer Institute, Kampala, Uganda
| | | | - Barnabas Atwiine
- Department of Pediatrics and Child Health, Mbarara University of Science and Technology, Head of Department, Mbarara, Uganda
| | - Ruth Namazzi
- Department of Pediatrics and Child Health, Makerere University College of Health Sciences, Head of Hematology-oncology, Kampala, Uganda
| | - Victor Musiime
- Department of Pediatrics and Child Health, Makerere University College of Health Sciences, Professor of Pediatrics, Kampala, Uganda
| | | | - Jaques van Heerden
- Department of pediatric Oncology, Antwerp University Hospital, Consultant Pediatric Oncologist, Antwerp, Belgium
| |
Collapse
|
15
|
Feng L, Yang X, Lu X, Kan Y, Wang C, Sun D, Zhang H, Wang W, Yang J. 18F-FDG PET/CT-based radiomics nomogram could predict bone marrow involvement in pediatric neuroblastoma. Insights Imaging 2022; 13:144. [PMID: 36057694 PMCID: PMC9440965 DOI: 10.1186/s13244-022-01283-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/07/2022] [Indexed: 11/10/2022] Open
Abstract
Objective To develop and validate an 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT)-based radiomics nomogram for non-invasively prediction of bone marrow involvement (BMI) in pediatric neuroblastoma. Methods A total of 133 patients with neuroblastoma were retrospectively included and randomized into the training set (n = 93) and test set (n = 40). Radiomics features were extracted from both CT and PET images. The radiomics signature was developed. Independent clinical risk factors were identified using the univariate and multivariate logistic regression analyses to construct the clinical model. The clinical-radiomics model, which integrated the radiomics signature and the independent clinical risk factors, was constructed using multivariate logistic regression analysis and finally presented as a radiomics nomogram. The predictive performance of the clinical-radiomics model was evaluated by receiver operating characteristic curves, calibration curves and decision curve analysis (DCA). Results Twenty-five radiomics features were selected to construct the radiomics signature. Age at diagnosis, neuron-specific enolase and vanillylmandelic acid were identified as independent predictors to establish the clinical model. In the training set, the clinical-radiomics model outperformed the radiomics model or clinical model (AUC: 0.924 vs. 0.900, 0.875) in predicting the BMI, which was then confirmed in the test set (AUC: 0.925 vs. 0.893, 0.910). The calibration curve and DCA demonstrated that the radiomics nomogram had a good consistency and clinical utility. Conclusion The 18F-FDG PET/CT-based radiomics nomogram which incorporates radiomics signature and independent clinical risk factors could non-invasively predict BMI in pediatric neuroblastoma. Supplementary Information The online version contains supplementary material available at 10.1186/s13244-022-01283-8.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Xu Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Xia Lu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Ying Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Chao Wang
- Sinounion Medical Technology (Beijing) Co., Ltd., Beijing, 100192, China
| | - Dehui Sun
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China
| | - Hui Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China.
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, 100050, China.
| |
Collapse
|
16
|
Seo ES, Shin M, Lim H, Cho HW, Ju HY, Cho YS, Yoo KH, Koo HH, Lee JW, Sung KW. Clinical implication of residual MIBG-positive disease in the follow-up of high-risk neuroblastoma treated with tandem high-dose chemotherapy and autologous stem cell transplantation. Pediatr Blood Cancer 2022; 69:e29502. [PMID: 34889513 DOI: 10.1002/pbc.29502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/24/2021] [Accepted: 11/09/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND The implication of residual metaiodobenzylguanidine (MIBG)-positive disease in the era of tandem high-dose chemotherapy (HDCT) with autologous stem cell transplantation (auto-SCT) has not yet been established in neuroblastoma. Moreover, most published studies have not evaluated the long-term prognosis of patients with residual MIBG-positive disease following treatment completion. Therefore, we investigated the prognostic significance of residual MIBG-positive disease at each treatment phase and after treatment completion. METHODS We assessed MIBG scans labeled with either iodine-123 (123 I) or 131 I from 150 patients with MIBG-avid and high-risk neuroblastoma enrolled in the NB-2004, -2009, and -2014 trials at postinduction, posttandem HDCT/auto-SCT, and completion of treatment. RESULTS The residual MIBG-positive disease at postinduction and posttandem HDCT/auto-SCT evaluation was highly correlated with the risk of progression. However, at treatment completion, there was no significant difference in survival and risk of progression between patients with residual MIBG-positive disease and MIBG-negative patients. Patients with persistent MIBG-positive disease at the end of treatment were more likely to have indolent tumor characteristics, such as favorable histology at diagnosis, lower incidence of MYCN amplification, and slow response to chemotherapy. CONCLUSION Residual MIBG-positive disease during treatment predicted unfavorable outcomes for patients with high-risk neuroblastoma, even under tandem HDCT/auto-SCT. However, persistent MIBG uptake at the completion of all treatments may not always indicate an active disease.
Collapse
Affiliation(s)
- Eun Seop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Muheon Shin
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hana Lim
- 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
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hong Hoe Koo
- 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
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Ladumor Y, Seong BKA, Hallett R, Valencia-Sama I, Adderley T, Wang Y, Kee L, Gont A, Kaplan DR, Irwin MS. Vitamin D Receptor Activation Attenuates Hippo Pathway Effectors and Cell Survival in Metastatic Neuroblastoma. Mol Cancer Res 2022; 20:895-908. [PMID: 35190818 DOI: 10.1158/1541-7786.mcr-21-0425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 01/12/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022]
Abstract
Survival for high-risk neuroblastoma remains poor. Most patients who recur, present with metastatic disease, and few targetable pathways that govern spread to distant sites are currently known. We previously developed a metastatic mouse model to select cells with enhanced ability to spread to the bone and brain and identified a signature based on differentially expressed genes, which also predicted patient survival. To discover new neuroblastoma therapies, we utilized the Connectivity Map to identify compounds that can reverse this metastatic transcriptional signature and found calcipotriol, a vitamin D3 analog, to be a compound that selectively targets cell lines with enhanced metastatic potential. Calcipotriol treatment of enhanced metastatic, but not parental, cells reduces proliferation and survival via vitamin D receptor (VDR) signaling, increases the expression of RASSF2, a negative regulator of the Hippo signaling pathway, and reduces the levels of the Hippo pathway effectors YAP and TAZ. RASSF2 is required for the effects of calcipotriol and for the reduction of levels and nuclear localization of YAP/TAZ. Migration of the enhanced metastatic cells and YAP/TAZ levels are reduced after calcipotriol treatment and YAP overexpression reduces calcipotriol sensitivity. Furthermore, metastatic cells that overexpress VDR also showed lower tumor burden in vivo. IMPLICATIONS This newly identified link between VDR signaling and the Hippo pathway could inform treatment strategies for metastatic neuroblastoma.
Collapse
Affiliation(s)
- Yagnesh Ladumor
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Bo Kyung Alex Seong
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Robin Hallett
- Cell Biology, Hospital for Sick Children, Toronto, Canada.,Neurosciences and Mental Health Programs, Hospital for Sick Children, Toronto, Canada
| | | | | | - Yingying Wang
- Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Lynn Kee
- Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Alexander Gont
- Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - David R Kaplan
- Neurosciences and Mental Health Programs, Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics and University of Toronto, Toronto, Canada
| | - Meredith S Irwin
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Cell Biology, Hospital for Sick Children, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada
| |
Collapse
|
18
|
Razmara AM, Wittenburg LA, Al-Nadaf S, Toedebusch RG, Meyers FJ, Toedebusch CM. Prevalence and Clinicopathologic Features of Canine Metastatic Melanoma Involving the Central Nervous System: A Retrospective Analysis and Comparative Review. Front Oncol 2022; 12:868004. [PMID: 35692802 PMCID: PMC9186031 DOI: 10.3389/fonc.2022.868004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Background Central nervous system (CNS) involvement is the leading cause of death in malignant melanoma. Rodent models, while vital to mechanistic investigation, have had limited success identifying effective therapies for melanoma brain metastases. The companion dog with de novo melanoma is a promising complementary model for developmental therapeutic investigation, as these tumors occur in an immunologically outbred host that has shared environmental exposures with humans. However, relatively little is known regarding the prevalence and clinicopathological features of canine melanoma metastasis to the CNS. To further validate the dog as an appropriate model for human metastatic melanoma, the aims of this study were to determine the rate of CNS metastasis and associated clinicopathologic features in canine malignant melanoma. Methods Medical records of dogs diagnosed with malignant melanoma from 1985-2019 at the University of California Davis Veterinary Medical Teaching Hospital were assessed retrospectively. Clinicopathologic features were compared between dogs with CNS metastasis (CNS+) and dogs without CNS metastasis (CNS-). Site of CNS involvement and associated neurological signs were analyzed via Wilcoxon-Mann-Whitney rank sum and Fisher’s exact tests. Survival data were analyzed via Kaplan-Meier estimates. Results CNS metastasis was identified in 38% of dogs in this study (20/53). The oral cavity was the most common site of primary melanoma in both groups [CNS+: n=12 (60%) vs. CNS-: n=22 (67%); p>0.99]. The total burden of metastatic disease was higher in the CNS+ group (CNS+: 4, 95% CI 3-5 vs. CNS-: 3, 95% CI 1-3; p<0.001). The cerebrum was the most common site of CNS metastasis (n=15, 75%) and seizures were the most observed neurological sign (n=9, 64%). There was no difference in overall survival between CNS+ and CNS- groups. However, the median survival time following onset of neurological signs was 9.5 days (95% CI 1-43), with 5 dogs euthanized within 24 hours of the onset of neurological signs. Conclusions Canine and human MM patients share similar rates of CNS metastasis and clinical presentation. This study will guide clinical management of canines with malignant melanoma and inform future studies using dogs with spontaneously occurring melanoma as a preclinical model for human melanoma brain metastases.
Collapse
Affiliation(s)
- Aryana M. Razmara
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- UCD Comprehensive Cancer Center, Sacramento, CA, United States
| | - Luke A. Wittenburg
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- UCD Comprehensive Cancer Center, Sacramento, CA, United States
| | - Sami Al-Nadaf
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- UCD Comprehensive Cancer Center, Sacramento, CA, United States
| | - Ryan G. Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- UCD Comprehensive Cancer Center, Sacramento, CA, United States
| | - Frederick J. Meyers
- UCD Comprehensive Cancer Center, Sacramento, CA, United States
- Department of Internal Medicine, Division of Hematology and Oncology, Center for Precision Medicine, University of California, Davis School of Medicine, Sacramento, CA, United States
| | - Christine M. Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- UCD Comprehensive Cancer Center, Sacramento, CA, United States
- *Correspondence: Christine M. Toedebusch,
| |
Collapse
|
19
|
Thomas Lucas J. Risk Factors Associated with Metastatic Site Failure in Patients with High-Risk Neuroblastoma. Clin Transl Radiat Oncol 2022; 34:42-50. [PMID: 35345864 PMCID: PMC8956847 DOI: 10.1016/j.ctro.2022.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 11/29/2022] Open
Abstract
We observed an increased hazard for failure at metastatic sites which remain persistently avid on MIBG following systemic therapy. -Limited response to induction therapy described by Curie and SIOPEN score selects patients at greater risk for poly-metastatic site failure. -The low proportion of metastatic sites treated with radiotherapy precluded definitive testing of its impact on the hazard for metastatic site failure. -Patients who are unable to undergo transplant, and/or have extensive disease at diagnosis (lung metastases) may be poor candidates for consolidative metastatic site directed radiotherapy given the high competing risk of failure at a new metastatic site.
Purpose This retrospective study sought to identify predictors of metastatic site failure (MSF) at new and/or original (present at diagnosis) sites in high-risk neuroblastoma patients. Methods and materials Seventy-six high-risk neuroblastoma patients treated on four institutional prospective trials from 1997 to 2014 with induction chemotherapy, surgery, myeloablative chemotherapy, stem-cell rescue, and were eligible for consolidative primary and metastatic site (MS) radiotherapy were eligible for study inclusion. Computed-tomography and I123 MIBG scans were used to assess disease response and Curie scores at diagnosis, post-induction, post-transplant, and treatment failure. Outcomes were described using the Kaplan–Meier estimator. Cox proportional hazards frailty (cphfR) and CPH regression (CPHr) were used to identify covariates predictive of MSF at a site identified either at diagnosis or later. Results MSF occurred in 42 patients (55%). Consolidative MS RT was applied to 30 MSs in 10 patients. Original-MSF occurred in 146 of 383 (38%) nonirradiated and 18 of 30 (60%) irradiated MSs (p = 0.018). Original- MSF occurred in postinduction MIBG-avid MSs in 68 of 81 (84%) nonirradiated and 12 of 14 (85%) radiated MSs (p = 0.867). The median overall and progression-free survival rates were 61 months (95% CI 42.6Not Reached) and 24.1 months (95% CI 16.538.7), respectively. Multivariate CPHr identified inability to undergo transplant (HR 32.4 95%CI 9.396.8, p < 0.001) and/or maintenance chemotherapy (HR 5.2, 95%CI 1.716.2, p = 0.005), and the presence of lung metastases at diagnosis (HR 4.4 95%CI 1.711.1, p = 0.002) as predictors of new MSF. The new MSF-free survival rate at 3 years was 25% and 87% in patients with and without high-risk factors. Conclusions Incremental improvements in systemic therapy influence the patterns and type of metastatic site failure in neuroblastoma. Persistence of MIBG-avidity following induction chemotherapy and transplant at MSs increased the hazard for MSF.
Collapse
|
20
|
Tas ML, Molenaar JJ, Peek AM, Lequin MH, Verdijk RM, de Krijger RR, Tytgat GA, van Noesel MM. Refractory Stage M Ganglioneuroblastoma With Bone Metastases and a Favorable, Chronic Course of Disease: Description of a Patient Cohort. J Pediatr Hematol Oncol 2022; 44:e5-e13. [PMID: 33885033 PMCID: PMC8728760 DOI: 10.1097/mph.0000000000002067] [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: 11/08/2020] [Accepted: 12/13/2020] [Indexed: 11/26/2022]
Abstract
Refractory stage M neuroblastoma (NB) is associated with a poor prognosis and a progressive course of disease. Here, we describe a unique group of patients with a discrepant clinical course. Seven histologically confirmed ganglioneuroblastoma (GNB) (n=6) and differentiating NB (n=1) patients were identified who were diagnosed with stage M disease based on iodine-123-metaiodobenzylguanidine avid bone metastases. Six patients started on high-risk treatment, without tumor response (stable disease). Treatment was discontinued before the start of consolidation treatment because of refractory response in all patients. Unexpectedly, after cessation of treatment no progression of disease occurred. In 2 patients, the primary tumors expanded (>25%) very slowly during 1.5 and 3 years, and remained stable thereafter. Metabolically, a slow decrease of urinary homovanillic acid and vanillylmandelic acid levels and iodine-123-metaiodobenzylguanidine avidity was observed. All patients are alive with presence of metastatic disease after a median follow-up of 17 years (range: 6.7 to 27 y). Interestingly, at diagnosis, 6 patients were asymptomatic, 6 patients had GNB morphology, and 5 patients had meningeal metastases. These are all features seen in only a small minority of stage M patients. This GNB entity illustrates the clinical heterogeneity of neuroblastic tumors and can be used to further study the developmental origin of different NB subtypes.
Collapse
Affiliation(s)
| | | | - Annemarie M.L. Peek
- Departments of Solid Tumors
- Department of Pediatric Oncology, Beatrix Children’s Hospital, University Medical Center Groningen, Groningen
| | - Maarten H. Lequin
- Departments of Solid Tumors
- Departments of Radiology and Nuclear Medicine
| | - Rob M. Verdijk
- Department of Pathology, Section Neuropathology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ronald R. de Krijger
- Diagnostics and Pathology, Princess Máxima Center for Pediatric Oncology
- Pathology, University Medical Center Utrecht, Utrecht
| | | | | |
Collapse
|
21
|
Retrospective Analysis of INRG Clinical and Genomic Factors for 605 Neuroblastomas in Japan: A Report from the Japan Children’s Cancer Group Neuroblastoma Committee (JCCG-JNBSG). Biomolecules 2021; 12:biom12010018. [PMID: 35053166 PMCID: PMC8774029 DOI: 10.3390/biom12010018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/31/2022] Open
Abstract
Neuroblastomas (NBs) exhibit broad and divergent clinical behaviors and tumor risk classification at diagnosis is crucial for the selection of an appropriate therapeutic strategy for each patient. The present study aimed to validate the clinical relevance of International Neuroblastoma Risk Group (INRG) prognostic and genomic markers in a Japanese NB cohort using a retrospective analysis. Follow-up data based on 30 common INRG queries in 605 NB cases diagnosed in Japan between 1990 and 2014 were collected and the genome signature of each tumor sample was integrated. As previously indicated, age, tumor stage, MYCN, DNA ploidy, the adrenals as the primary tumor site, serum ferritin and lactate dehydrogenase (LDH) levels, segmental chromosome aberrations, and the number of chromosome breakpoints (BP) correlated with lower survival rates, while the thorax as the primary tumor site and numerical chromosome aberrations correlated with a favorable prognosis. In the patient group with stage 4, MYCN non-amplified tumors (n = 225), one of the challenging subsets for risk stratification, age ≥ 18 months, LDH ≥ 1400 U/L, and BP ≥ 7 correlated with lower overall and event-free survival rates (p < 0.05). The genome subgroup GG-P2s (partial chromosome gain/loss type with 1p/11q losses and 17q gain, n = 30) was strongly associated with a lower overall survival rate (5-year survival rate: 34%, p < 0.05). Therefore, the combination of the tumor genomic pattern (GG-P2s and BP ≥ 7) with age at diagnosis and LDH will be a promising predictor for MYCN-non-amplified high-risk NBs in patient subsets, in accordance with previous findings from the INRG project.
Collapse
|
22
|
Shawraba F, Hammoud H, Mrad Y, Saker Z, Fares Y, Harati H, Bahmad HF, Nabha S. Biomarkers in Neuroblastoma: An Insight into Their Potential Diagnostic and Prognostic Utilities. Curr Treat Options Oncol 2021; 22:102. [PMID: 34580780 DOI: 10.1007/s11864-021-00898-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2021] [Indexed: 12/23/2022]
Abstract
OPINION STATEMENT Neuroblastoma (NB) is a heterogeneous solid tumor of the pediatric population that originates from neural crest cells and affects the developing sympathetic nervous system. It is the most common neuroblastic tumor accounting for approximately 10% of all childhood cancers and 10-15% of pediatric tumor mortalities. The outcomes range from spontaneous tumor regression in low-risk groups to metastasis and death even after multimodal therapy in high-risk groups. Hence, the detection of NB at an early stage improves outcomes and provides a better prognosis for patients. Early detection and prognosis of NB depend on specific molecules termed biomarkers which can be tissue-specific or circulating. Certain biomarkers are employed in the classification of NB into different groups to improve the treatment and prognosis, and others can be used as therapeutic targets. Therefore, novel biomarker discovery is essential for the early detection of NB, predicting the course of the disease, and developing new targeted treatment strategies. In this review, we aim to summarize the literature pertinent to some important biomarkers of NB and discuss the prognostic role of these biomarkers as well as their potential role in targeted therapy.
Collapse
Affiliation(s)
- Fatima Shawraba
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Hussein Hammoud
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Yara Mrad
- Université Clermont Auvergne, Inserm, Neuro-Dol, Clermont-Ferrand, France
| | - Zahraa Saker
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Youssef Fares
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon.,Department of Neurosurgery, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Hisham F Bahmad
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, 4300 Alton Rd, Miami Beach, FL, 33140, USA.
| | - Sanaa Nabha
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon.
| |
Collapse
|
23
|
Woo S, Araji A, El Amine MA, Gangai N, Acquafredda E, Price AP, Trippett TM, Hricak H, Vargas HA, Behr GG. Emergency room imaging in pediatric patients with cancer: analysis of the spectrum and frequency of imaging modalities and findings in a tertiary cancer center and their relationship with survival. Cancer Imaging 2021; 21:51. [PMID: 34454626 PMCID: PMC8400759 DOI: 10.1186/s40644-021-00421-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 08/16/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To assess the spectrum and frequency of modalities used for emergency room (ER) imaging and their findings in pediatric cancer patients and assess their relationship with survival. METHODS Consecutive pediatric cancer patients that underwent imaging during an ER visit at our tertiary cancer center over a 5-year period were retrospectively analyzed. Imaging findings were considered positive when they were relevant to the ER presenting complaint. Imaging positivity was correlated with inpatient admission. Overall survival (OS) was assessed with Kaplan-Meier curves and uni- and multi-variate Cox proportional hazards model was used to identify significant factors associated with OS. RESULTS Two hundred sixty-one patients (135 males and 126 females; median age 11 years [interquartile range 5-16 years] with 348 visits and a total of 406 imaging studies were included. Common chief complaints were related to the chest (100 [28.7 %]) and fever (99 [28.4 %]). ER imaging was positive in 207 visits (59.5 %), commonly revealing increased metastases (50 [14.4 %]), pneumonia (47 [13.5 %]), and other lung problems (12 [2.9 %]). Positive ER imaging was associated with inpatient admission (69.3 % [133/192] vs. 40.4 % [63/156], p < 0.01). Multivariate survival analysis showed that positive ER imaging (hazard ratio [HR] = 2.35 [95% CI 1.44-3.83, p < 0.01), admission (HR = 1.86 [95% CI 1.17-3.00], p < 0.01), number of ER visits (HR = 3.08 [95% CI 1.62-5.83], p < 0.01 for ≥ 3 visits) were associated with poorer survival. CONCLUSIONS Imaging was able to delineate the cause for ER visits in children with cancer in over half of the cases. Positive ER imaging was associated with admission and worse survival.
Collapse
Affiliation(s)
- Sungmin Woo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA.
| | - Abdallah Araji
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| | - Mohammad Ali El Amine
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| | - Natalie Gangai
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| | - Elizabeth Acquafredda
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| | - Anita P Price
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| | - Tanya M Trippett
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hedvig Hricak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| | - Hebert Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| | - Gerald G Behr
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, 10065, New York, NY, USA
| |
Collapse
|
24
|
Liquid biomarkers for the management of paediatric neuroblastoma: an approach to personalised and targeted cancer therapy. JOURNAL OF RADIOTHERAPY IN PRACTICE 2021. [DOI: 10.1017/s1460396920000102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
AbstractBackground:Neuroblastoma is the most common extracranial solid tumour of infancy and accounts for about 6–10% of paediatric cancers. It has a biologically and clinically heterogeneous behaviour that ranges from spontaneous regression to cases of highly aggressive metastatic disease that could be unresponsive to standard therapy. In recent years, there have been several investigations into the development of various diagnostic, predictive and prognostic biomarkers towards personalised and targeted management of the disease.Materials and Methods:This paper reports on the review of current clinical and emerging biomarkers used in risk assessment, screening for early detection and diagnosis, prognostication and monitoring of the response of treatment of neuroblastoma in paediatric patients.Conclusions:Tumour markers can significantly improve diagnosis; however, the invasive, unpleasant and inconvenient nature of current tissue biopsies limits their applications, especially in paediatric patients. Therefore, the development of a non-invasive, reliable high accurate and personalised diagnostic tool capable of early detection and rapid response is the most promising step towards advanced cancer management from tumour diagnosis, therapy to patient monitoring and represents an important step towards the promise of precision, personalised and targeted medicine. Liquid biopsy assay with wide ranges of clinical applications is emerging to hold incredible potential for advancing cancer treatment and has greater promise for diagnostic purposes, identification and tracking of tumour-specific alterations during the course of the disease and to guide therapeutic decisions.
Collapse
|
25
|
Yamazaki F, Yamasaki K, Kiyotani C, Hashii Y, Shioda Y, Hara J, Matsumoto K. Thiotepa-melphalan myeloablative therapy for high-risk neuroblastoma. Pediatr Blood Cancer 2021; 68:e28896. [PMID: 33788375 DOI: 10.1002/pbc.28896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 12/02/2020] [Accepted: 12/26/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND Appropriate high-dose chemotherapy (HDC) for high-risk neuroblastoma has not yet been established. In Japan, a unique HDC regimen that comprises two cycles of a total of 800 mg/m2 of thiotepa and a total of 280 mg/m2 of melphalan is widely utilized. METHODS To evaluate the safety and efficacy of this thiotepa-melphalan high-dose therapy for high-risk neuroblastoma, we reviewed the medical records of 41 patients with high-risk neuroblastoma who underwent this regimen followed by autologous peripheral blood stem cell rescue between 2002 and 2012. MYCN-amplified high-risk neuroblastomas were observed in 23 patients. All patients underwent intensive multidrug induction chemotherapy, but none underwent anti-GD2 antibody immunotherapy. The primary tumor was resected at the adequate time point. RESULTS The median follow-up duration for living patients was 9.2 years (range 5.5-14.0 years). The 5-year event-free survival (EFS) and overall survival from treatment initiation were 41.5 ± 7.7% and 56.1 ± 7.8%, respectively. The 5-year EFS of MYCN-amplified high-risk neuroblastoma patients was 60.9 ± 10.2%, which was significantly superior compared with those with MYCN-nonamplified high-risk neuroblastoma (16.7 ± 8.8%; p < .001). MYCN amplification was the most favorable prognostic factor for EFS (hazard ratio = 0.29; 95% confidence interval = 0.12-0.66). Of the 41 patients, three died because of regimen-related toxicity (infection, n = 2; microangiopathy, n = 1). CONCLUSION The thiotepa-melphalan high-dose therapy with thiotepa and melphalan may be effective for high-risk neuroblastoma. However, this regimen is toxic and warrants special attention in clinical practice.
Collapse
Affiliation(s)
- Fumito Yamazaki
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan.,Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Kai Yamasaki
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Chikako Kiyotani
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan
| | - Yoshiko Hashii
- Department of Pediatrics, Osaka University Hospital, Osaka, Japan
| | - Yoko Shioda
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan
| | - Junichi Hara
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Kimikazu Matsumoto
- National Center for Child Health and Development, Children's Cancer Center, Tokyo, Japan
| |
Collapse
|
26
|
He B, Mao J, Huang L. Clinical Characteristics and Survival Outcomes in Neuroblastoma With Bone Metastasis Based on SEER Database Analysis. Front Oncol 2021; 11:677023. [PMID: 34141621 PMCID: PMC8203907 DOI: 10.3389/fonc.2021.677023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/11/2021] [Indexed: 12/29/2022] Open
Abstract
Purpose Clinical features and survival analysis of neuroblastoma (NB) are well explored. However, clinical research of NB patients with bone metastasis is rarely reported. Thus, the current study was performed to analyze the clinical features, survival outcome, and risk factors in those patients. Materials and Methods We reviewed the Surveillance, Epidemiology, and End Results (SEER) database to select cases diagnosed with NB with bone metastasis from 2010 to 2016. Overall survival (OS) and cancer-specific survival (CSS) were analyzed through univariate Cox regression analysis. Subsequently, we performed multivariate analysis to determine independent predictors of survival. The Kaplan–Meier method was applied to intuitively show differences in prognostic value between independent risk factors. Results We finally identified 393 NB patients with bone metastasis who were selected for survival analysis. Nearly half of the patients (47.3%) were aged >3 years. The adrenal gland was the primary tumor site, accounting for approximately two thirds of cases (66.2%). The 5-year OS and CSS rates of all patients were 62.1% and 64.1%, respectively. The univariate analysis indicated that age, lung metastasis, and tumor size were significantly associated with OS and CSS. Based on the multivariable analysis, age at 2 and 3 years, lung metastasis, and tumor size >10 cm remained significant negative predictors of OS and CSS. Conclusion For NB patients with bone metastasis, three independent prognostic risk factors (age, lung metastasis, and tumor size) are helpful to clinicians for predicting prognosis and guiding treatment. Reasonable treatment modalities for these patients should be further investigated to prolong survival.
Collapse
Affiliation(s)
- Bin He
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China.,Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, China
| | - Jianshui Mao
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Leyi Huang
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
27
|
Samim A, Tytgat GA, Bleeker G, Wenker ST, Chatalic KL, Poot AJ, Tolboom N, van Noesel MM, Lam MG, de Keizer B. Nuclear Medicine Imaging in Neuroblastoma: Current Status and New Developments. J Pers Med 2021; 11:jpm11040270. [PMID: 33916640 PMCID: PMC8066332 DOI: 10.3390/jpm11040270] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/01/2021] [Indexed: 12/20/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid malignancy in children. At diagnosis, approximately 50% of patients present with metastatic disease. These patients are at high risk for refractory or recurrent disease, which conveys a very poor prognosis. During the past decades, nuclear medicine has been essential for the staging and response assessment of neuroblastoma. Currently, the standard nuclear imaging technique is meta-[123I]iodobenzylguanidine ([123I]mIBG) whole-body scintigraphy, usually combined with single-photon emission computed tomography with computed tomography (SPECT-CT). Nevertheless, 10% of neuroblastomas are mIBG non-avid and [123I]mIBG imaging has relatively low spatial resolution, resulting in limited sensitivity for smaller lesions. More accurate methods to assess full disease extent are needed in order to optimize treatment strategies. Advances in nuclear medicine have led to the introduction of radiotracers compatible for positron emission tomography (PET) imaging in neuroblastoma, such as [124I]mIBG, [18F]mFBG, [18F]FDG, [68Ga]Ga-DOTA peptides, [18F]F-DOPA, and [11C]mHED. PET has multiple advantages over SPECT, including a superior resolution and whole-body tomographic range. This article reviews the use, characteristics, diagnostic accuracy, advantages, and limitations of current and new tracers for nuclear medicine imaging in neuroblastoma.
Collapse
Affiliation(s)
- Atia Samim
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Godelieve A.M. Tytgat
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
| | - Gitta Bleeker
- Department of Radiology and Nuclear Medicine, Northwest Clinics, Wilhelminalaan 12, 1815 JD Alkmaar, The Netherlands;
| | - Sylvia T.M. Wenker
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Kristell L.S. Chatalic
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Alex J. Poot
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Nelleke Tolboom
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Max M. van Noesel
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
| | - Marnix G.E.H. Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
| | - Bart de Keizer
- Princess Maxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, The Netherlands; (A.S.); (G.A.M.T.); (S.T.M.W.); (K.L.S.C.); (A.J.P.); (N.T.); (M.M.v.N.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands;
- Correspondence: ; Tel.: +31-887-571-794
| |
Collapse
|
28
|
Van Heerden J, Kruger M, Esterhuizen TM, Hendricks M, Du Plessis J, Engelbrecht G, Janse van Vuuren M, van Emmenes B, Uys R, Burger C, Nyakale N, More S, Brink A. The Association between Tumour Markers and Meta-iodobenzylguanidine Scans in South African Children with High-risk Neuroblastoma. Clin Oncol (R Coll Radiol) 2021; 33:517-526. [PMID: 33781675 DOI: 10.1016/j.clon.2021.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/04/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
AIMS Diagnostic and post-induction 123I-meta-iodobenzylguanidine (123I-mIBG) scans have prognostic significance in the treatment of neuroblastoma, but data from low- and middle-income countries are limited due to resource constraints. The aim of this study was to determine the association between neuroblastoma-associated tumour markers (lactate dehydrogenase [LDH], ferritin and MYCN amplification) and 123I-mIBG scans (modified Curie scores and metastatic disease patterns) in predicting complete metastatic response rates (mCR) and overall survival. MATERIALS AND METHODS Two hundred and ninety patients diagnosed with high-risk neuroblastoma in South Africa between January 2000 and May 2018 and a subanalysis of 78 patients with diagnostic 123I-mIBG scans were included. Data collection included LDH, ferritin and MYCN amplification at diagnosis. Two nuclear physicians independently determined the modified Curie scores and pattern of distribution for each diagnostic and post-induction 123I-mIBG scans with high inter-rater agreement (r = 0.952) and reliability (K = 0.805). The cut-off values for the diagnostic and post-induction modified Curie scores of ≥7.0 (P = 0.026) and 3 (P = 0.009), respectively, were generated. The association between the tumour markers and the modified Curie score of the 123I-mIBG scans was determined using post-induction mCR and 2-year overall survival. RESULTS Diagnostic LDH (P < 0.001), ferritin (P < 0.001) and the diagnostic modified Curie scores (P = 0.019) significantly predicted mCR. Only ferritin correlated with diagnostic modified Curie scores (P = 0.003) but had a low correlation coefficient of 0.353. On multivariable analysis, the only significant covariate for 2-year overall survival at diagnosis was LDH <750 U/l (P = 0.024). A post-induction chemotherapy modified Curie score ≤3.0 had a 2-year overall survival of 46.2% compared with 30.8% for a score >3.0 (P = 0.484). CONCLUSION LDH, ferritin and the diagnostic 123I-mIBG scans significantly predicted mCR, but only LDH predicted 2-year overall survival. Ferritin and the modified Curie scores correlated with each other. MYCN amplification neither correlated with any aspect of the 123I-mIBG scans nor significantly predicted mCR or 2-year overall survival. LDH and ferritin are therefore appropriate neuroblastoma tumour markers to be used in low- and middle-income countries with limited or no access to mIBG scans and/or MYCN amplification studies.
Collapse
Affiliation(s)
- J Van Heerden
- Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa; Paediatric Haematology and Oncology, Department of Paediatrics, Antwerp University Hospital, Antwerp, Belgium.
| | - M Kruger
- Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - T M Esterhuizen
- Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - M Hendricks
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of Cape Town, Paediatric Haematology and Oncology Service, Red Cross War Memorial Children's Hospital, Cape Town, South Africa
| | - J Du Plessis
- Department of Paediatrics, Faculty of Health Sciences, University of the Free State, Division of Paediatric Haematology and Oncology, Universitas Hospital, Bloemfontein, South Africa
| | - G Engelbrecht
- Department of Nuclear Medicine, University of the Free State, Universitas Hospital, Bloemfontein, South Africa
| | - M Janse van Vuuren
- Drs B Vorster and M Janse van Vuuren Incorporated, Nuclear Physicians, Bloemfontein, South Africa
| | - B van Emmenes
- Division of Paediatric Haematology and Oncology Hospital, Department of Paediatrics, Frere Hospital, East London, South Africa
| | - R Uys
- Paediatric Haematology and Oncology, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - C Burger
- Department of Nuclear Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa
| | - N Nyakale
- Department of Nuclear Medicine, Faculty of Health Sciences, University of KwaZulu-Natal, Inkosi Albert Luthuli Academic Hospital, Durban, South Africa
| | - S More
- Department of Paediatrics and Child Health, Division of Nuclear Medicine, Faculty of Health Sciences, University of Cape Town, Red Cross War Memorial Children's Hospital, Cape Town, South Africa
| | - A Brink
- Department of Paediatrics and Child Health, Division of Nuclear Medicine, Faculty of Health Sciences, University of Cape Town, Red Cross War Memorial Children's Hospital, Cape Town, South Africa
| |
Collapse
|
29
|
Pulmonary Metastasis of Low-risk Perinatal Neuroblastoma After Resection: Implications for Surveillance. J Pediatr Hematol Oncol 2021; 43:e184-e186. [PMID: 31815890 DOI: 10.1097/mph.0000000000001693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/08/2019] [Indexed: 11/25/2022]
Abstract
In the wake of the Children's Oncology Group (COG) ANBL00P2 trial and the ongoing ANBL1232 trial, an increasing number of children with neonatal neuroblastoma are being managed nonoperatively. We report the case of a patient with low-risk, non-MYCN amplified, neuroblastoma that was diagnosed and resected in the neonatal period but subsequently developed pulmonary metastases by the age of 7 months. Though rare, the possibility of low-risk disease metastasizing during surveillance should be recognized and may not be identified by current protocols.
Collapse
|
30
|
Moreno L, Guo D, Irwin MS, Berthold F, Hogarty M, Kamijo T, Morgenstern D, Pasqualini C, Ash S, Potschger U, Ladenstein R, Valteau-Couanet D, Cohn SL, Pearson ADJ, London WB. A nomogram of clinical and biologic factors to predict survival in children newly diagnosed with high-risk neuroblastoma: An International Neuroblastoma Risk Group project. Pediatr Blood Cancer 2021; 68:e28794. [PMID: 33205902 DOI: 10.1002/pbc.28794] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/16/2020] [Accepted: 10/09/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Long-term outcome remains poor for children with high-risk neuroblastoma (five-year overall survival [OS] ∼50%). Our objectives were to (a) identify prognostic biomarkers and apply them in a nomogram to identify the subgroup of ultra-high-risk patients at highest risk of disease progression/death, for whom novel frontline therapy is urgently needed; and (b) validate the nomogram in an independent cohort. METHODS A total of 1820 high-risk patients (≥18 months old with metastatic neuroblastoma), diagnosed 1998-2015, from the International Neuroblastoma Risk Groups (INRG) Data Commons were analyzed in a retrospective cohort study. Using multivariable Cox regression of OS from diagnosis, a nomogram was created from prognostic biomarkers to predict three-year OS. External validation was performed using the SIOPEN HR-NBL1 trial cohort (n = 521), evidenced by receiver operating characteristic curves. RESULTS The nomogram, including MYCN status (P < 0.0001), lactate dehydrogenase (LDH) (P = 0.0007), and presence of bone marrow metastases (P = 0.004), had robust performance and was validated. Applying the nomogram at diagnosis (a) gives prognosis of an individual patient and (b) identifies patients predicted to have poor outcome (three-year OS was 30% ± 5% for patients with a nomogram score of > 82 points; 58% ± 1% for those ≤82 points). Median follow-up time was 5.5 years (range, 0-14.1). CONCLUSIONS In high-risk neuroblastoma, a novel, publicly available nomogram using prognostic biomarkers (MYCN status, LDH, presence of bone marrow metastases; https://neuroblastoma.shinyapps.io/High-Risk-Neuroblastoma-Nomogram/) has the flexibility to apply a clinically suitable and context-specific cutoff to identify patients at highest risk of death. This will facilitate testing urgently needed new frontline treatment options to improve outcome for these children.
Collapse
Affiliation(s)
- Lucas Moreno
- Pediatric Hematology and Oncology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Dongjing Guo
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Meredith S Irwin
- Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Frank Berthold
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Michael Hogarty
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, Pennsylvania
| | - Takehiko Kamijo
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Daniel Morgenstern
- Department of Paediatrics, Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | | | - Shifra Ash
- Pediatric Hematology Oncology Center, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | | | | | | | - Susan L Cohn
- Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Andrew D J Pearson
- Division of Clinical Studies, Institute of Cancer Research, Royal Marsden Hospital, Sutton, UK
| | - Wendy B London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
31
|
Morandi F, Sabatini F, Podestà M, Airoldi I. Immunotherapeutic Strategies for Neuroblastoma: Present, Past and Future. Vaccines (Basel) 2021; 9:43. [PMID: 33450862 PMCID: PMC7828327 DOI: 10.3390/vaccines9010043] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 12/22/2022] Open
Abstract
Neuroblastoma is the most common extracranial pediatric solid tumor with a heterogeneous clinical course, ranging from spontaneous regression to metastatic disease and death, irrespective of intensive chemotherapeutic regimen. On the basis of several parameters, children affected by neuroblastoma are stratified into low, intermediate and high risk. At present, more than 50% of high-risk patients with metastatic spread display an overall poor long-term outcome also complicated by devastating long-term morbidities. Thus, novel and more effective therapies are desperately needed to improve lifespan of high-risk patients. In this regard, adoptive cell therapy holds great promise and several clinical trials are ongoing, demonstrating safety and tolerability, with no toxicities. Starting from the immunological and clinical features of neuroblastoma, we here discuss the immunotherapeutic approaches currently adopted for high-risk patients and different innovative therapeutic strategies currently under investigation. The latter are based on the infusion of natural killer (NK) cells, as support of consolidation therapy in addition to standard treatments, or chimeric antigen receptor (CAR) T cells directed against neuroblastoma associated antigens (e.g., disialoganglioside GD2). Finally, future perspectives of adoptive cell therapies represented by γδ T lymphocyes and CAR NK cells are envisaged.
Collapse
Affiliation(s)
| | | | | | - Irma Airoldi
- Laboratorio Cellule Staminali Post-Natali e Terapie Cellulari, Istituto Giannina Gaslini (Istituto di Ricerca e Cura a Carattere Scientifico—IRCCS), Via G. Gaslini 5, 16147 Genova, Italy; (F.M.); (F.S.); (M.P.)
| |
Collapse
|
32
|
Jazmati D, Butzer S, Hero B, Doyen J, Ahmad Khalil D, Steinmeier T, Schulze Schleithoff S, Eggert A, Simon T, Timmermann B. Long-term follow-up of children with neuroblastoma receiving radiotherapy to metastatic lesions within the German Neuroblastoma Trials NB97 and NB 2004. Strahlenther Onkol 2020; 197:683-689. [PMID: 33300099 PMCID: PMC8292260 DOI: 10.1007/s00066-020-01718-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Neuroblastoma (NB) is the most common extracranial solid malignancy during childhood. Despite a multimodal treatment approach, the prognosis of patients with metastatic NB is not satisfactory. Although radiotherapy (RT) has become an integral part of treatment of the primary tumor, the role of RT in osteomedullary lesions is not well defined. A retrospective analysis was conducted to evaluate the impact of RT for metastatic sites in children with high-risk NB. METHODS All patients with stage 4 NB from the prospective, multicenter NB trials NB97 and NB2004 who received RT to metastatic sites during frontline treatment were included in this retrospective analysis. RESULTS A total of 18 children were irradiated with a median dose of 36 Gray (Gy; range 20-45 Gy) to one or more (range 1-3) osteomedullary metastases with or without concomitant RT to the primary tumor site. The median follow-up time was 149 months (range 55-220) in survivors. At 5 years, local relapse-free survival (LRFS) at irradiated metastatic sites and metastases-free survival (MFS) at distant, non-irradiated site rates were 51.4 and 39.9%, respectively. The estimated overall survival (OS) rate at 5 years was 49.4%. No high-grade acute or late toxicity and no secondary malignancy was reported. CONCLUSION RT to metastases is feasible for patients with stage 4 NB. However, an impact of RT to residual metastatic sites on outcome was not found. Studies with larger cohorts or prospective trials would be desirable in order to elucidate the role of RT for metastases.
Collapse
Affiliation(s)
- Danny Jazmati
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| | - Sarina Butzer
- Children's Hospital, University of Cologne, Cologne, Germany
| | - Barbara Hero
- Children's Hospital, University of Cologne, Cologne, Germany
| | - Jerome Doyen
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Dalia Ahmad Khalil
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Theresa Steinmeier
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Stefanie Schulze Schleithoff
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Angelika Eggert
- Department of Pediatrics, Division of Oncology and Hematology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Thorsten Simon
- Children's Hospital, University of Cologne, Cologne, Germany
| | - Beate Timmermann
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| |
Collapse
|
33
|
Campos Cogo S, Gradowski Farias da Costa do Nascimento T, de Almeida Brehm Pinhatti F, de França Junior N, Santos Rodrigues B, Regina Cavalli L, Elifio-Esposito S. An overview of neuroblastoma cell lineage phenotypes and in vitro models. Exp Biol Med (Maywood) 2020; 245:1637-1647. [PMID: 32787463 PMCID: PMC7802384 DOI: 10.1177/1535370220949237] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
This review was conducted to present the main neuroblastoma (NB) clinical characteristics and the most common genetic alterations present in these pediatric tumors, highlighting their impact in tumor cell aggressiveness behavior, including metastatic development and treatment resistance, and patients' prognosis. The distinct three NB cell lineage phenotypes, S-type, N-type, and I-type, which are characterized by unique cell surface markers and gene expression patterns, are also reviewed. Finally, an overview of the most used NB cell lines currently available for in vitro studies and their unique cellular and molecular characteristics, which should be taken into account for the selection of the most appropriate model for NB pre-clinical studies, is presented. These valuable models can be complemented by the generation of NB reprogrammed tumor cells or organoids, derived directly from patients' tumor specimens, in the direction toward personalized medicine.
Collapse
Affiliation(s)
- Sheron Campos Cogo
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | | | | | - Nilton de França Junior
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | - Bruna Santos Rodrigues
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| | - Luciane Regina Cavalli
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, Brazil
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Selene Elifio-Esposito
- Graduate Program in Health Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil
| |
Collapse
|
34
|
Abstract
Informative and realistic mouse models of high-risk neuroblastoma are central to understanding mechanisms of tumour initiation, progression, and metastasis. They also play vital roles in validating tumour drivers and drug targets, as platforms for assessment of new therapies and in the generation of drug sensitivity data that can inform treatment decisions for individual patients. This review will describe genetically engineered mouse models of specific subsets of high-risk neuroblastoma, the development of patient-derived xenograft models that more broadly represent the diversity and heterogeneity of the disease, and models of primary and metastatic disease. We discuss the research applications, advantages, and limitations of each model type, the importance of model repositories and data standards for supporting reproducible, high-quality research, and potential future directions for neuroblastoma mouse models.
Collapse
Affiliation(s)
- Alvin Kamili
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Caroline Atkinson
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Toby N Trahair
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia.,School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia. .,School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
35
|
Seo ES, Lee EJ, Lee B, Shin M, Cho YS, Hyun JK, Cho HW, Ju HY, Yoo KH, Koo HH, Lee JW, Sung KW. Metastatic Burden Defines Clinically and Biologically Distinct Subgroups of Stage 4 High-Risk Neuroblastoma. J Clin Med 2020; 9:jcm9092730. [PMID: 32847064 PMCID: PMC7565784 DOI: 10.3390/jcm9092730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
This study aimed to identify the prognostic subgroups of stage 4 high-risk neuroblastoma based on metastatic burden and explore their distinct clinical and genomic features. Patients aged ≥18 months with stage 4 and metaiodobenzylguanidine-avid neuroblastoma were enrolled. One hundred and thirty eligible patients were treated under the tandem high-dose chemotherapy scheme. Prognostic significance of metastatic burden measured by the modified Curie score was analyzed using a competing risk approach, and the optimal cut-point was determined. Metastasis-specific subgroups (cut-point: 26) were compared using clinicopathological variables, and differential gene expression analysis and gene set variation analysis (GSVA) were performed using RNA sequencing (RNA-seq). Metastatic burden at diagnosis showed a progressive association with relapse/progression. After applying the cut-point, patients with high metastatic burden showed >3-fold higher risk of relapse/progression than those with low metastatic burden. Moreover, patients with high metastatic burden showed smaller primary tumors and higher biochemical marker levels than those with low metastatic burden. In the genomic analysis, 51 genes were found to be differentially expressed based on the set criteria. GSVA revealed 55 gene sets, which significantly distinguished patients with high metastatic burden from those with low metastatic burden at a false discovery rate <0.25. The results indicated the prognostic significance of metastatic burden in stage 4 high-risk neuroblastoma, and we identified the distinct clinicopathological and genomic features based on metastatic burden. This study may aid in the better understanding and risk-stratification of stage 4 high-risk neuroblastoma patients.
Collapse
Affiliation(s)
- Eun Seop Seo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
| | - Eun-jin Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (E.-j.L.); (B.L.)
| | - Boram Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (E.-j.L.); (B.L.)
| | - Muheon Shin
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (M.S.); (Y.-S.C.)
| | - Young-Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (M.S.); (Y.-S.C.)
| | - Ju Kyung Hyun
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
| | - Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
| | - Hee Young Ju
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
- Correspondence: (J.W.L.); (K.W.S.); Tel.: +82-2-3410-0659 (J.W.L.); +82-2-3410-3529 (K.W.S.); Fax: +82-2-3410-0049 (K.W.S.)
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (E.S.S.); (J.K.H.); (H.W.C.); (H.Y.J.); (K.H.Y.); (H.H.K.)
- Correspondence: (J.W.L.); (K.W.S.); Tel.: +82-2-3410-0659 (J.W.L.); +82-2-3410-3529 (K.W.S.); Fax: +82-2-3410-0049 (K.W.S.)
| |
Collapse
|
36
|
Chen D, Cox J, Annam J, Weingart M, Essien G, Rathi KS, Rokita JL, Khurana P, Cuya SM, Bosse KR, Pilgrim A, Li D, Shields C, Laur O, Maris JM, Schnepp RW. LIN28B promotes neuroblastoma metastasis and regulates PDZ binding kinase. Neoplasia 2020; 22:231-241. [PMID: 32339949 PMCID: PMC7186370 DOI: 10.1016/j.neo.2020.04.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/28/2020] [Accepted: 04/01/2020] [Indexed: 12/17/2022]
Abstract
Neuroblastoma is an aggressive pediatric malignancy of the neural crest with suboptimal cure rates and a striking predilection for widespread metastases, underscoring the need to identify novel therapeutic vulnerabilities. We recently identified the RNA binding protein LIN28B as a driver in high-risk neuroblastoma and demonstrated it promotes oncogenic cell proliferation by coordinating a RAN-Aurora kinase A network. Here, we demonstrate that LIN28B influences another key hallmark of cancer, metastatic dissemination. Using a murine xenograft model of neuroblastoma dissemination, we show that LIN28B promotes metastasis. We demonstrate that this is in part due to the effects of LIN28B on self-renewal and migration, providing an understanding of how LIN28B shapes the metastatic phenotype. Our studies reveal that the let-7 family, which LIN28B inhibits, decreases self-renewal and migration. Next, we identify PDZ Binding Kinase (PBK) as a novel LIN28B target. PBK is a serine/threonine kinase that promotes the proliferation and self-renewal of neural stem cells and serves as an oncogenic driver in multiple aggressive malignancies. We demonstrate that PBK is both a novel direct target of let-7i and that MYCN regulates PBK expression, thus elucidating two oncogenic drivers that converge on PBK. Functionally, PBK promotes self-renewal and migration, phenocopying LIN28B. Taken together, our findings define a role for LIN28B in neuroblastoma metastasis and define the targetable kinase PBK as a potential novel vulnerability in metastatic neuroblastoma.
Collapse
Affiliation(s)
- Dongdong Chen
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Julie Cox
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jayabhargav Annam
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Melanie Weingart
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Grace Essien
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Komal S Rathi
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Priya Khurana
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Selma M Cuya
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Kristopher R Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adeiye Pilgrim
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Daisy Li
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Cara Shields
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | | | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert W Schnepp
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
37
|
Berthold F, Faldum A, Ernst A, Boos J, Dilloo D, Eggert A, Fischer M, Frühwald M, Henze G, Klingebiel T, Kratz C, Kremens B, Krug B, Leuschner I, Schmidt M, Schmidt R, Schumacher-Kuckelkorn R, von Schweinitz D, Schilling FH, Theissen J, Volland R, Hero B, Simon T. Extended induction chemotherapy does not improve the outcome for high-risk neuroblastoma patients: results of the randomized open-label GPOH trial NB2004-HR. Ann Oncol 2020; 31:422-429. [PMID: 32067684 DOI: 10.1016/j.annonc.2019.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Long-term survival of high-risk neuroblastoma patients is still below 50% despite intensive multimodal treatment. This trial aimed to address whether the addition of two topotecan-containing chemotherapy courses compared to standard induction therapy improves event-free survival (EFS) of these patients. PATIENTS AND METHODS An open-label, multicenter, prospective randomized controlled trial was carried out at 58 hospitals in Germany and Switzerland. Patients aged 1-21 years with stage 4 neuroblastoma and patients aged 6 months to 21 years with MYCN-amplified tumors were eligible. The primary endpoint was EFS. Patients were randomly assigned to standard induction therapy with six chemotherapy courses or to experimental induction chemotherapy starting with two additional courses of topotecan, cyclophosphamide, and etoposide followed by standard induction chemotherapy (eight courses in total). After induction chemotherapy, all patients received high-dose chemotherapy with autologous hematopoietic stem cell rescue and isotretinoin for consolidation. Radiotherapy was applied to patients with active tumors at the end of induction chemotherapy. RESULTS Of 536 patients enrolled in the trial, 422 were randomly assigned to the control arm (n = 211) and the experimental arm (n = 211); the median follow-up time was 3.32 years (interquartile range 1.65-5.92). At data lock, the 3-year EFS of experimental and control patients was 34% and 32% [95% confidence Interval (CI) 28% to 40% and 26% to 38%; P = 0.258], respectively. Similarly, the 3-year overall survival of the patients did not differ [54% and 48% (95% CI 46% to 62% and 40% to 56%), respectively; P = 0.558]. The response to induction chemotherapy was not different between the arms. The median number of non-fatal toxicities per patient was higher in the experimental group while the median number of toxicities per chemotherapy course was not different. CONCLUSION While the burden for the patients was increased by prolonging the induction chemotherapy and the toxicity, the addition of two topotecan-containing chemotherapy courses did not improve the EFS of high-risk neuroblastoma patients and thus cannot be recommended. CLINICAL TRIALS. GOV NUMBER NCT number 03042429.
Collapse
Affiliation(s)
- F Berthold
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany.
| | - A Faldum
- Institute of Medical Statistics and Clinical Research, University of Muenster, Muenster, Germany
| | - A Ernst
- Institute of Medical Statistics and Computational Biology (IMSB), University of Cologne, Cologne, Germany
| | - J Boos
- Department of Pediatric Oncology and Hematology, University of Muenster, Muenster, Germany
| | - D Dilloo
- Department of Pediatric Oncology and Hematology, University of Bonn, Bonn, Germany
| | - A Eggert
- Department of Pediatric Oncology and Hematology, Charité Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
| | - M Fischer
- Department of Experimental Pediatric Oncology and Center for Molecular Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - M Frühwald
- Swabian Children's Cancer Center, Children's Hospital, University Hospital Augsburg, Augsburg, Germany
| | - G Henze
- Department of Pediatric Oncology and Hematology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - T Klingebiel
- Department of Children and Adolescents, University Hospital, Goethe University Frankfurt (Main), Frankfurt am Main, Germany
| | - C Kratz
- Department of Pediatric Oncology and Hematology, Medicinal University, Hannover, Germany
| | - B Kremens
- Department of Pediatric Oncology and Hematology, University of Essen, Essen, Germany
| | - B Krug
- Institute of Diagnostic and Interventional Radiology, University of Cologne, Cologne, Germany
| | - I Leuschner
- Children's Tumor Registry, Institute of Pathology, University of Kiel, Kiel, Germany
| | - M Schmidt
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - R Schmidt
- Institute of Medical Statistics and Clinical Research, University of Muenster, Muenster, Germany
| | | | - D von Schweinitz
- Department of Pediatric Surgery, University of Munich, Munich, Germany
| | - F H Schilling
- Department of Pediatric Oncology and Hematology, Olgahospital Stuttgart, Stuttgart, Germany
| | - J Theissen
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - R Volland
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - B Hero
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - T Simon
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| |
Collapse
|
38
|
Ornell KJ, Coburn JM. Developing preclinical models of neuroblastoma: driving therapeutic testing. BMC Biomed Eng 2019; 1:33. [PMID: 32903387 PMCID: PMC7422585 DOI: 10.1186/s42490-019-0034-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 11/19/2019] [Indexed: 12/14/2022] Open
Abstract
Despite advances in cancer therapeutics, particularly in the area of immuno-oncology, successful treatment of neuroblastoma (NB) remains a challenge. NB is the most common cancer in infants under 1 year of age, and accounts for approximately 10% of all pediatric cancers. Currently, children with high-risk NB exhibit a survival rate of 40–50%. The heterogeneous nature of NB makes development of effective therapeutic strategies challenging. Many preclinical models attempt to mimic the tumor phenotype and tumor microenvironment. In vivo mouse models, in the form of genetic, syngeneic, and xenograft mice, are advantageous as they replicated the complex tumor-stroma interactions and represent the gold standard for preclinical therapeutic testing. Traditional in vitro models, while high throughput, exhibit many limitations. The emergence of new tissue engineered models has the potential to bridge the gap between in vitro and in vivo models for therapeutic testing. Therapeutics continue to evolve from traditional cytotoxic chemotherapies to biologically targeted therapies. These therapeutics act on both the tumor cells and other cells within the tumor microenvironment, making development of preclinical models that accurately reflect tumor heterogeneity more important than ever. In this review, we will discuss current in vitro and in vivo preclinical testing models, and their potential applications to therapeutic development.
Collapse
Affiliation(s)
- Kimberly J Ornell
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA 01605 USA
| | - Jeannine M Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA 01605 USA
| |
Collapse
|
39
|
Manenq C, Lesesve JF, Dreumont N, Massin F, Salignac S, Mansuy L, Chastagner P, Latger-Cannard V, Broséus J. Combined use of multiparametric flow cytometry and cytomorphology to enhance detection of neuroblastoma metastatic cells in bone marrow. Int J Lab Hematol 2019; 42:52-60. [PMID: 31821742 DOI: 10.1111/ijlh.13137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/02/2019] [Accepted: 11/18/2019] [Indexed: 02/02/2023]
Abstract
INTRODUCTION In the context of neuroblastoma (NB), the screening for bone marrow (BM) metastasis is a recurrent issue for hematology laboratory routine practice. Detection of low tumor burden using light microscopy is often difficult. In this regard, our objective was to evaluate the performance of multiparametric flow cytometry (FC) for detecting NB metastatic cells in BM. METHODS We applied a new FC multiparametric panel allowing the analysis of the co-expression of 5 surface markers: GD2 (disialoganglioside 2), CD9, CD56, CD81, and CD90, on CD45-negative BM cell populations, and compared results with BM biopsy immunohistochemistry, which is the reference method. RESULTS In spike-in tests, the multiparametric FC successfully detected NB cells mixed in peripheral blood mononuclear cells to a level of 0.01%. FC analysis was performed on 45 sets of BM aspirates sampled from 21 children, either at diagnosis or during follow-up. Combining multiparametric FC with light microscopy improved NB metastasis detection, with a higher sensitivity (76.9% vs 61.5%) and a higher specificity (94.4% vs 77.8%) as compared to light microscopy alone. At the time of diagnosis, multiparametric FC detected NB metastatic cells in all cases. CONCLUSION These results illustrate the performance of multiparametric FC analysis to detect metastatic BM infiltration of NB. This is of particular interest in an emergency context, since when combined with light microscopy, it enhances the detection of metastatic invasion within a short timeframe, allowing an adapted and rapid clinical management.
Collapse
Affiliation(s)
- Christine Manenq
- Université de Lorraine, CHRU-Nancy, Service d'Hématologie Biologique, Pôle Laboratoires, France.,Université de Lorraine, CHRU-Nancy, Plate-forme de Cytométrie en Flux, Pôle Laboratoires, France
| | - Jean-François Lesesve
- Université de Lorraine, CHRU-Nancy, Service d'Hématologie Biologique, Pôle Laboratoires, France.,Université de Lorraine, CHRU-Nancy, Plate-forme de Cytométrie en Flux, Pôle Laboratoires, France.,Université de Lorraine, Inserm, NGERE, Nancy, France
| | | | - Frédéric Massin
- Université de Lorraine, CHRU-Nancy, Plate-forme de Cytométrie en Flux, Pôle Laboratoires, France
| | - Sylvain Salignac
- Université de Lorraine, CHRU-Nancy, Service d'Hématologie Biologique, Pôle Laboratoires, France.,Université de Lorraine, CHRU-Nancy, Plate-forme de Cytométrie en Flux, Pôle Laboratoires, France
| | - Ludovic Mansuy
- Université de Lorraine, CHRU-Nancy, Service d'Oncologie Pédiatrique, Pôle Enfants Néonatalogie, France
| | - Pascal Chastagner
- Université de Lorraine, CHRU-Nancy, Service d'Oncologie Pédiatrique, Pôle Enfants Néonatalogie, France
| | - Véronique Latger-Cannard
- Université de Lorraine, CHRU-Nancy, Service d'Hématologie Biologique, Pôle Laboratoires, France.,Université de Lorraine, CHRU-Nancy, Plate-forme de Cytométrie en Flux, Pôle Laboratoires, France
| | - Julien Broséus
- Université de Lorraine, CHRU-Nancy, Service d'Hématologie Biologique, Pôle Laboratoires, France.,Université de Lorraine, CHRU-Nancy, Plate-forme de Cytométrie en Flux, Pôle Laboratoires, France.,Université de Lorraine, Inserm, NGERE, Nancy, France
| |
Collapse
|
40
|
Odone-Filho V, Cristofani LM, Maluf PT, Almeida MTA, Halley N, Vince CSC, de Azambuja AMP, Brumatti M, Lubraico P, da Camara Lopes LHA, Leite KRM, Silva JLF, Plese JPP, Weltman E. Involvement of the central nervous system in neuroblastomas: A potential direct pathway. Med Hypotheses 2019; 136:109479. [PMID: 31778890 DOI: 10.1016/j.mehy.2019.109479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/25/2019] [Accepted: 11/08/2019] [Indexed: 11/24/2022]
Abstract
Although frequently disseminated to other anatomical sites, neuroblastoma (NB) is rarely reported as involving the central nervous system (CNS), which may reflect insufficient research in poorly controlled systemic disease. Here we demonstrate the involvement of the CNS in patients with NB over 18 months of age at diagnosis of extensive systemic disease. Meningeal metastases were observed even in the presence of complete systemic control. Although no improvement in patient's survival was observed, radiotherapy was effective in preventing CNS recurrence after observation of actual or previous dural disease. In conclusion, this study uncovered the uncommon pathologic involvement of the CNS in children with advanced NB and underscores the meningeal surface as a potential pathway for this to occur.
Collapse
Affiliation(s)
- Vicente Odone-Filho
- Sao Paulo University Medical School, Pediatric Department, Hematology-Oncology Division, Brazil.
| | - Lilian Maria Cristofani
- Sao Paulo University Medical School, Pediatric Department, Hematology-Oncology Division, Brazil
| | - Paulo Taufi Maluf
- Sao Paulo University Medical School, Pediatric Department, Hematology-Oncology Division, Brazil
| | | | - Nathalia Halley
- Hematology-Oncology Division, Pediatric Department, Sao Paulo University Medical School, Brazil
| | | | | | - Melina Brumatti
- Hematology-Oncology Division, Pediatric Department, Sao Paulo University Medical School, Brazil
| | - Priscilla Lubraico
- Hematology-Oncology Division, Pediatric Department, Sao Paulo University Medical School, Brazil
| | | | | | | | | | - Eduardo Weltman
- Hospital Israelita Albert Einstein, Department of Radiotherapy, Brazil
| |
Collapse
|
41
|
Bagatell R, Irwin MS. Tandem Transplant for High-Risk Neuroblastoma: Next Steps in the Era of Precision Medicine. JAMA 2019; 322:729-731. [PMID: 31454025 DOI: 10.1001/jama.2019.11641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Rochelle Bagatell
- Perelman School of Medicine, Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia
| | - Meredith S Irwin
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Patel HD, Singla N, Ghandour RA, Freifeld Y, Cheaib JG, Woldu SL, Pierorazio PM, Bagrodia A. Site of extranodal metastasis impacts survival in patients with testicular germ cell tumors. Cancer 2019; 125:3947-3952. [PMID: 31355922 DOI: 10.1002/cncr.32427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/28/2019] [Accepted: 06/26/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Using a large, nationally representative, population-based cancer registry, this study systematically evaluated the impact of the location and burden of extranodal testicular germ cell tumor (TGCT) metastases on survival. METHODS Men with stage III TGCTs captured by the Surveillance, Epidemiology, and End Results registry from 2010 to 2015 with distant extranodal metastases were identified. Clinicopathologic information was collected, and patients were subdivided according to the specific organ site or sites of metastatic involvement (lung, liver, bone, and/or brain). Kaplan-Meier analysis and multivariable Cox regression were used to evaluate cancer-specific survival (CSS), and model performance was assessed with Harrell's C statistic. RESULTS Nine hundred sixty-nine patients with stage III TGCTs were included with predominantly nonseminomatous histology (84%). Most patients (91%) had pulmonary metastases, whereas 20%, 10%, and 10% had liver, bone, and brain metastases, respectively. Over a median follow-up of 21 months, 19% of these men died of TGCTs. When they were grouped by the primary site of metastasis, patients with more than 1 extrapulmonary metastasis exhibited the worst CSS (hazard ratio [HR] vs isolated pulmonary involvement, 4.27; 95% confidence interval [CI], 2.60-7.00; P < .01). Among patients with isolated extrapulmonary involvement, those with brain metastases had the poorest survival (HR, 3.24; 95% CI, 1.98-5.28; P < .01), and they were followed by patients with liver (HR, 2.29; 95% CI, 1.56-3.35; P < .01) and bone metastases (HR, 1.97; 95% CI, 1.11-3.50; P = .02). Harrell's C statistic (multivariable) was 0.71. CONCLUSIONS The site of metastatic involvement affects survival outcomes for patients with TGCTs, and this may reflect both the aggressive biology and the challenging treatment of these tumors. Further incorporation of organotropism into current prognostic models for metastatic TGCTs warrants attention.
Collapse
Affiliation(s)
- Hiten D Patel
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nirmish Singla
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Rashed A Ghandour
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Yuval Freifeld
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Joseph G Cheaib
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Solomon L Woldu
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| | - Phillip M Pierorazio
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern, Dallas, Texas
| |
Collapse
|
43
|
Trigg RM, Shaw JA, Turner SD. Opportunities and challenges of circulating biomarkers in neuroblastoma. Open Biol 2019; 9:190056. [PMID: 31088252 PMCID: PMC6544987 DOI: 10.1098/rsob.190056] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Molecular analysis of nucleic acid and protein biomarkers is becoming increasingly common in paediatric oncology for diagnosis, risk stratification and molecularly targeted therapeutics. However, many current and emerging biomarkers are based on analysis of tumour tissue, which is obtained through invasive surgical procedures and in some cases may not be accessible. Over the past decade, there has been growing interest in the utility of circulating biomarkers such as cell-free nucleic acids, circulating tumour cells and extracellular vesicles as a so-called liquid biopsy of cancer. Here, we review the potential of emerging circulating biomarkers in the management of neuroblastoma and highlight challenges to their implementation in the clinic.
Collapse
Affiliation(s)
- Ricky M. Trigg
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Jacqui A. Shaw
- Leicester Cancer Research Centre, College of Life Sciences, University of Leicester, Leicester LE2 7LX, UK
| | - Suzanne D. Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
44
|
Peinemann F, van Dalen EC, Enk H, Tytgat GAM, Cochrane Childhood Cancer Group. Anti-GD2 antibody-containing immunotherapy postconsolidation therapy for people with high-risk neuroblastoma treated with autologous haematopoietic stem cell transplantation. Cochrane Database Syst Rev 2019; 4:CD012442. [PMID: 31016728 PMCID: PMC6479178 DOI: 10.1002/14651858.cd012442.pub2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neuroblastoma is a rare malignant disease that primarily affects children. The tumours mainly develop in the adrenal medullary tissue, and an abdominal mass is the most common presentation. High-risk disease is characterised by metastasis and other primary tumour characteristics resulting in increased risk for an adverse outcome. The GD2 carbohydrate antigen is expressed on the cell surface of neuroblastoma tumour cells and is thus a promising target for anti-GD2 antibody-containing immunotherapy. OBJECTIVES To assess the efficacy of anti-GD2 antibody-containing postconsolidation immunotherapy after high-dose chemotherapy (HDCT) and autologous haematopoietic stem cell transplantation (HSCT) compared to standard therapy after HDCT and autologous HSCT in people with high-risk neuroblastoma. Our primary outcomes were overall survival and treatment-related mortality. Our secondary outcomes were progression-free survival, event-free survival, early toxicity, late non-haematological toxicity, and health-related quality of life. SEARCH METHODS We searched the electronic databases CENTRAL (2018, Issue 9), MEDLINE (PubMed), and Embase (Ovid) on 20 September 2018. We searched trial registries and conference proceedings on 28 October 2018. Further searches included reference lists of recent reviews and relevant articles as well as contacting experts in the field. There were no limits on publication year or language. SELECTION CRITERIA Randomised controlled trials evaluating anti-GD2 antibody-containing immunotherapy after HDCT and autologous HSCT in people with high-risk neuroblastoma. DATA COLLECTION AND ANALYSIS Two review authors independently performed study selection, abstracted data on study and participant characteristics, and assessed risk of bias and GRADE. Any differences were resolved by discussion, with third-party arbitration unnecessary. We performed analyses according to the guidelines of the Cochrane Handbook for Systematic Reviews of Interventions. We used the five GRADE considerations, that is study limitations, consistency of effect, imprecision, indirectness, and publication bias, to judge the quality of the evidence. MAIN RESULTS We identified one randomised controlled trial that included 226 people with high-risk neuroblastoma who were pre-treated with autologous HSCT. The study randomised 113 participants to receive immunotherapy including isotretinoin, granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-2, and ch14.18, a type of anti-GD2 antibody also known as dinutuximab. The study randomised another 113 participants to receive standard therapy including isotretinoin.The results on overall survival favoured the dinutuximab-containing immunotherapy group (hazard ratio (HR) 0.50, 95% confidence interval (CI) 0.31 to 0.80; P = 0.004). The results on event-free survival also favoured the dinutuximab-containing immunotherapy group (HR 0.61, 95% CI 0.41 to 0.92; P = 0.020). Randomised data on adverse events were not reported separately. The study did not report progression-free survival, late non-haematological toxicity, and health-related quality of life as separate endpoints. We graded the quality of the evidence as moderate. AUTHORS' CONCLUSIONS The evidence base favours dinutuximab-containing immunotherapy compared to standard therapy concerning overall survival and event-free survival in people with high-risk neuroblastoma pre-treated with autologous HSCT. Randomised data on adverse events are lacking, therefore more research is needed before definitive conclusions can be made regarding this outcome.
Collapse
Affiliation(s)
- Frank Peinemann
- Children's Hospital, University of ColognePediatric Oncology and HematologyKerpener Str. 62CologneGermany50937
| | - Elvira C van Dalen
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25UtrechtNetherlands3584 CS
| | - Heike Enk
- c/o Cochrane Childhood CancerAmsterdamNetherlands
| | - Godelieve AM Tytgat
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25UtrechtNetherlands3584 CS
| | | |
Collapse
|
45
|
Morgenstern DA, Bagatell R, Cohn SL, Hogarty MD, Maris JM, Moreno L, Park JR, Pearson AD, Schleiermacher G, Valteau-Couanet D, London WB, Irwin MS. The challenge of defining "ultra-high-risk" neuroblastoma. Pediatr Blood Cancer 2019; 66:e27556. [PMID: 30479064 DOI: 10.1002/pbc.27556] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/08/2018] [Accepted: 10/27/2018] [Indexed: 12/17/2022]
Abstract
Given the biological and clinical heterogeneity of neuroblastoma, risk stratification is vital to determining appropriate treatment. Historically, most patients with high-risk neuroblastoma (HR-NBL) have been treated uniformly without further stratification. Attempts have been made to identify factors that can be used to risk stratify these patients and to characterize an "ultra-high-risk" (UHR) subpopulation with particularly poor outcome. However, among published data, there is a lack of consensus in the definition of the UHR population and heterogeneity in the endpoints and statistical methods used. This review summarizes our current understanding of stratification of HR-NBL and discusses the complex issues in defining UHR neuroblastoma.
Collapse
Affiliation(s)
| | - Rochelle Bagatell
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | | | - Michael D Hogarty
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - John M Maris
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Lucas Moreno
- Hospital Universitario Niño Jesus, Madrid, Spain
| | - Julie R Park
- Seattle Children's Hospital and University of Washington School of Medicine, Seattle, Washington
| | - Andrew D Pearson
- Institute of Cancer Research and Royal Marsden National Health Service (NHS) Foundation Trust, Sutton, Surrey, UK
| | | | | | - Wendy B London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, Massachusetts
| | - Meredith S Irwin
- Hospital for Sick Children and University of Toronto, Toronto, Canada
| |
Collapse
|
46
|
Morgenstern DA, Pötschger U, Moreno L, Papadakis V, Owens C, Ash S, Pasqualini C, Luksch R, Garaventa A, Canete A, Elliot M, Wieczorek A, Laureys G, Kogner P, Malis J, Ruud E, Beck-Popovic M, Schleiermacher G, Valteau-Couanet D, Ladenstein R. Risk stratification of high-risk metastatic neuroblastoma: A report from the HR-NBL-1/SIOPEN study. Pediatr Blood Cancer 2018; 65:e27363. [PMID: 30015396 DOI: 10.1002/pbc.27363] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/18/2018] [Accepted: 06/23/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Risk stratification is crucial to treatment decision-making in neuroblastoma. This study aimed to explore factors present at diagnosis affecting outcome in patients aged ≥18 months with metastatic neuroblastoma and to develop a simple risk score for prognostication. PROCEDURE Data were derived from the European high-risk neuroblastoma 1 (HR-NBL1)/International Society for Paediatric Oncology European Neuroblastoma (SIOPEN) trial with analysis restricted to patients aged ≥18 months with metastatic disease and treated prior to the introduction of immunotherapy. Primary endpoint was 5-year event-free survival (EFS). Prognostic factors assessed were sex, age, tumour MYCN amplification (MNA) status, serum lactate dehydrogenase (LDH)/ferritin, primary tumour and metastatic sites. Factors significant in univariate analysis were incorporated into a multi-variable model and an additive scoring system developed based on estimated log-cumulative hazard ratios. RESULTS The cohort included 1053 patients with median follow-up 5.5 years and EFS 27 ± 1%. In univariate analyses, age; serum LDH and ferritin; involvement of bone marrow, bone, liver or lung; and >1 metastatic system/compartment were associated with worse EFS. Tumour MNA was not associated with worse EFS. A multi-variable model and risk score incorporating age (>5 years, 2 points), serum LDH (>1250 U/L, 1 point) and number of metastatic systems (>1, 2 points) were developed. EFS was significantly correlated with risk score: EFS 52 ± 9% for score = 0 versus 6 ± 3% for score = 5 (P < 0.0001). CONCLUSIONS A simple score can identify an "ultra-high risk" (UHR) cohort (score = 5) comprising 8% of patients with 5-year EFS <10%. These patients appear not to benefit from induction therapy and could potentially be directed earlier to alternative experimental therapies in future trials.
Collapse
Affiliation(s)
- Daniel A Morgenstern
- Paediatric Haematology/Oncology, Hospital for Sick Children and University of Toronto, Toronto, Canada.,Paediatric Haematology/Oncology, Great Ormond Street Hospital and UCL Institute of Child Health, London, UK
| | - Ulrike Pötschger
- Studies and Statistics on Integrated Research and Projects, St Anna Kinderkrebsforschung, Vienna, Austria
| | - Lucas Moreno
- Paediatric Haematology/Oncology, Hospital Nino Jesús, Madrid, Spain
| | - Vassilios Papadakis
- Paediatric Haematology/Oncology, Agia Sofia Children's Hospital, Athens, Greece
| | - Cormac Owens
- Paediatric Haematology/Oncology, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - Shifra Ash
- Paediatric Haematology/Oncology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Claudia Pasqualini
- Department of Paediatric and Adolescent Oncology, Institut Gustav Roussy, Viellejuif, France
| | - Roberto Luksch
- Dipartimento di Ematologia e Onco-ematologia Pediatrica, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Adela Canete
- Pediatric Oncology Unit, Hospital Universitario y Politecnico La Fe, Valencia, Spain
| | - Martin Elliot
- Paediatric Oncology, Leeds Teaching Hospital NHS Trust, Leeds, UK
| | - Aleksandra Wieczorek
- Department of Pediatric Oncology and Hematology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Geneviève Laureys
- Department of Pediatric Hematology/Oncology and Stem Cell Transplantation University Hospital Ghent, Ghent, Belgium
| | - Per Kogner
- Department of Women's and Children's Health, Karolinska Institutet, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Josef Malis
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Prague, Czech Republic
| | - Ellen Ruud
- Department of Paediatric Medicine, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Maja Beck-Popovic
- Department of Pediatrics and Pediatric Surgery, Pediatric Haematology Oncology Unit, University Hospital Lausanne, Lausanne, Switzerland
| | | | | | - Ruth Ladenstein
- Paediatric Haematology/Oncology, St Anna Kinderspital and St Anna Kinderkrebforschung, Vienna, Austria
| |
Collapse
|
47
|
Ma Y, Zheng J, Feng J, Chen L, Dong K, Xiao X. Neuroblastomas in Eastern China: a retrospective series study of 275 cases in a regional center. PeerJ 2018; 6:e5665. [PMID: 30245940 PMCID: PMC6148411 DOI: 10.7717/peerj.5665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/29/2018] [Indexed: 01/17/2023] Open
Abstract
Purpose Most studies on neuroblastoma (NB) have been conducted in Western countries or Japan. The objective of our study was to analyze clinical and pathological features, MYCN status, surgical methods, and prognosis in Chinese NB patients. Methods A retrospective, single-center case series study of 275 NBs was implemented. Clinical manifestations, pathological features, MYCN status, and surgical treatment were analyzed. Log-rank test and Cox hazards models were used to assess overall survivals (OSs). Results The cohort consisted of 105 females and 170 males, with an age range of five days to 15 years. MYCN amplification was detected in 21.5% of all cases. The median OS was 15.0 months for MYCN amplified group. The five-year OS rates were 70.8% and 18.3% for MYCN unamplified and amplified groups, respectively, and the comparison of Kaplan-Meier curves for these two groups showed statistical significance (P < .001 by log-rank test). Gross total resection (GTR, n = 111) and subtotal resection (STR, n = 58) were administered in 169 patients at stages 3 and 4 who received chemotherapy and the comparison of Kaplan-Meier curves for different groups in these patients had statistical significance (STR vs. GTR, P = .009; MYCN unamplified vs. amplified, P < .001 by log-rank test, respectively).The multivariate survival analyses showed statistical significance (STR vs. GTR, P = .047; MYCN unamplified vs. amplified, P = .001 by Cox regression model). Conclusions MYCN amplification is an independently adverse prognostic factor in Chinese NB patients at stages 3 and 4 and GTR is associated with improved OS compared with STR in these patients.
Collapse
Affiliation(s)
- Yangyang Ma
- Department of Pathology, Children's Hospital of Fudan University, Shanghai, China
| | - Jicui Zheng
- Department of Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Jiayan Feng
- Department of Pathology, Children's Hospital of Fudan University, Shanghai, China
| | - Lian Chen
- Department of Pathology, Children's Hospital of Fudan University, Shanghai, China
| | - Kuiran Dong
- Department of Surgery, Children's Hospital of Fudan University, Shanghai, China
| | - Xianmin Xiao
- Department of Surgery, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
48
|
Fletcher JI, Ziegler DS, Trahair TN, Marshall GM, Haber M, Norris MD. Too many targets, not enough patients: rethinking neuroblastoma clinical trials. Nat Rev Cancer 2018; 18:389-400. [PMID: 29632319 DOI: 10.1038/s41568-018-0003-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuroblastoma is a rare solid tumour of infancy and early childhood with a disproportionate contribution to paediatric cancer mortality and morbidity. Combination chemotherapy, radiation therapy and immunotherapy remains the standard approach to treat high-risk disease, with few recurrent, actionable genetic aberrations identified at diagnosis. However, recent studies indicate that actionable aberrations are far more common in relapsed neuroblastoma, possibly as a result of clonal expansion. In addition, although the major validated disease driver, MYCN, is not currently directly targetable, multiple promising approaches to target MYCN indirectly are in development. We propose that clinical trial design needs to be rethought in order to meet the challenge of providing rigorous, evidence-based assessment of these new approaches within a fairly small patient population and that experimental therapies need to be assessed at diagnosis in very-high-risk patients rather than in relapsed and refractory patients.
Collapse
Affiliation(s)
- Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Toby N Trahair
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Glenn M Marshall
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia
- School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia.
- University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
49
|
Neuroblastoma: clinical and biological approach to risk stratification and treatment. Cell Tissue Res 2018; 372:195-209. [PMID: 29572647 DOI: 10.1007/s00441-018-2821-2] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/28/2018] [Indexed: 01/15/2023]
Abstract
Neuroblastoma is the most common extra-cranial solid tumor of childhood and the most common in the first year of life. It is a unique malignancy in that infants often present with either localized or metastatic disease that can spontaneously regress without intervention while older children can succumb to the disease after months to years of arduous therapy. Given this wide range of outcomes, the International Neuroblastoma Risk Group was created to stratify patients based on presenting characteristics and tumor biology in order to guide intensity of treatment strategies. The goal has been to decrease therapy for low-risk patients to avoid long-term complications while augmenting and targeting therapies for high-risk patients to improve overall survival. The international risk stratification depends on age, stage, histology, MYCN gene amplification status, tumor cell ploidy and segmental chromosomal abnormalities. Treatment for asymptomatic low-risk patients with an estimated survival of > 98% is often observation or surgical resection alone, whereas intermediate-risk patients with an estimated survival of > 90% require moderate doses of response-adjusted chemotherapy along with resection. High-risk patients undergo multiple cycles of combination chemotherapy before surgery, followed by consolidation with myeloablative autologous hematopoietic stem cell transplantation and local radiation and finally immunotherapy with differentiation therapy as maintenance phase. With this approach, outcome for patients with neuroblastoma has improved, as the field continues to expand efforts in more targeted therapies for high-risk patients.
Collapse
|
50
|
Tingting H, Di S, Xiaoping C, Xiaohong W, Dong H. High preoperative serum ferritin predicted poor prognosis in non-metastatic colorectal cancer. Saudi Med J 2017; 38:268-275. [PMID: 28251222 PMCID: PMC5387903 DOI: 10.15537/smj.2017.3.16110] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Objectives: To validate the prognostic significance of preoperative serum iron metabolism parameters in non-metastatic colorectal cancer patients treated with curative resection. Methods: We conducted a prospective cohort study in the Department of Surgical Oncology, WuXi 4th People’s Hospital, WuxiChina, between March 2010 and September 2013. The relationships of serum iron metabolism parameters with other variables were examined. The prognostic significance was evaluated using the Kaplan Meier curve and Cox proportional hazards regression model. Results: Five hundred and fourteen patients were eligible for analysis. The levels of the 3 iron metabolism parameters were interdependent. Hemoglobin level was positively correlated with serum iron and transferrin, and was negatively correlated with ferritin. Compared with peri-neural invasion (PNI)-negative patients, PNI-positive patients had higher serum iron (p=0.03) and ferritin levels (p=0.01). Compared with patients with the lowest quartile level of ferritin, patients with the highest quartile level of ferritin had a 2.21 (95% CI: 1.18-4.14) fold increased mortality risk in the univariate and 2.56 (95% CI: 1.10-5.96) in the multivariate Cox proportional hazards models. When stratified by TNM stages, it was only in stage III patients that serum ferritin remained statistically prognostically significant. Conclusions: Preoperative serum ferritin appeared as an independent adverse risk factor in non-metastatic colorectal cancer.
Collapse
Affiliation(s)
- Hong Tingting
- Department of Medical Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, People Republic of China. E-mail.
| | | | | | | | | |
Collapse
|